PROTEINS CONTAINING MULTIPLE, DIFFERENT UNNATURAL AMINO ACIDS AND METHODS OF MAKING AND USING SUCH PROTEINS

The invention relates generally to methods of producing a protein comprising a first unnatural amino acid (UAA) and a second, different UAA, and proteins comprising a first UAA and a second, different UAA.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of and priority to U.S. Provisional Patent Application No. 62/912,171, filed Oct. 8, 2019, which is incorporated herein by reference in its entirety for all purposes.

FIELD OF THE INVENTION

The present disclosure relates, in general, to the field where orthogonal tRNA/aminoacyl-tRNA synthetase pairs are used for the incorporation of multiple, different UAAs into a protein of interest as it is being synthesized.

BACKGROUND

In nature, proteins are produced in cells via processes known as transcription and translation. During transcription, a gene comprising a series of codons that collectively encode a protein of interest is transcribed into messenger RNA (mRNA). During translation, a ribosome, attaches to and moves along the mRNA and incorporates specific amino acids into a polypeptide chain being synthesized (translated) from the mRNA at positions corresponding to the codons to produce the protein. During translation specific, naturally occurring amino acids coupled to transfer RNAs (tRNAs) enter the ribosome. The tRNAs, which contain an anti-codon sequence, hybridize to their respective codon sequences in mRNA and transfer the amino acid they are carrying into the nascent protein chain at the appropriate position as the protein is synthesized.

Over the last few decades, significant efforts have been made to produce homogenous preparations of site-specifically modified proteins, e.g., mammalian proteins, on commercial scale quantities for use in a variety of applications, including, for example, therapeutics and diagnostics. Furthermore, efforts have been made to produce these modified mammalian proteins in eukaryotic cells (e.g., mammalian cells) because the proteins may be more readily produced in a properly folded and fully active form and/or post-translationally modified in a manner similar to the native protein naturally produced in a mammalian cell.

One approach for producing proteins that contain site-specific modifications involves the site-specific incorporation of one or more unnatural amino acids (UAAs) into a protein of interest. The ability to site-specifically incorporate UAAs into proteins in vivo has become a powerful tool to augment protein function or introduce new chemical functionalities not found in nature. The core elements required for this technology include: an engineered tRNA, an engineered aminoacyl-tRNA synthetase (aaRS) that charges the tRNA with a UAA, and a unique codon, e.g., a stop codon, directing the incorporation of the UAA into the protein as it is being synthesized.

Central to this approach is the use of an engineered tRNA/aaRS pair in which the aaRS charges the tRNA with the UAA of interest without cross-reacting with the tRNAs and amino acids normally present in the expression host cell. This has been accomplished by using an engineered tRNA/aaRS pair derived from an organism in different domain of life as the expression host cell so as to maximize the orthogonality between the engineered tRNA/aaRS pair (e.g., an engineered bacterial tRNA/aaRS pair) and the tRNA/aaRS pairs naturally found in the expression host cell (e.g., mammalian cell). The engineered tRNA, which is charged with the UAA via the aaRS, binds or hybridizes to the unique codon, such as a premature stop codon (UAG, UGA, UAA) present in the mRNA encoding the protein to be expressed. See, for example, FIG. 1, which shows the synthesis of a protein using an endogenous tRNA and an endogenous aaRS from the expression host cell and an engineered orthogonal tRNA and an orthogonal aaRS introduced into the host cell so as to facilitate the incorporation of a UAA into the protein as it is synthesized via the ribosome. To date, a variety of orthogonal tRNA/aaRS pairs have been produced for certain of the naturally occurring amino acids (see, e.g., U.S. Patent Publication US2017/0349891, and Zheng et al. (2018) BIOCHEM. 57:441-445). The approach facilitates the expression of proteins containing site specific modifications such as bioconjugation handles and photoactivatable crosslinkers, which can be used as therapeutics (e.g., antibody drug conjugates (ADCs), bi-specific antibodies (e.g., bispecific monoclonal antibodies), nanobodies, chemokines, vaccines, coagulation factors, hormones, and enzymes).

Despite the efforts made to date, there has been limited success in utilizing this technology to incorporate multiple, different UAAs into a single polypeptide. To incorporate multiple, different UAAs into one polypeptide, each UAA must be encoded by a distinct nonsense codon, and each UAA must be charged by different orthogonal aaRS/tRNA pairs, where each pair must also be orthogonal to each other (mutually orthogonal). A significant degree of cross-reactivity has been found between aaRS/tRNA pairs, which together with an inability to decode the full complement of nonsense codons has limited the overall efficiency of site-specific incorporation of two distinct UAAs in mammalian cells.

Accordingly, there remains a need for mammalian-based expression platforms that allow for the incorporation of multiple, distinct unnatural amino acids into proteins with high efficiency.

SUMMARY OF THE INVENTION

The invention is based, in part, on the discovery of combinations of nonsense codons, tRNAs, aminoacyl-tRNA synthetases, and UAAs that allow for efficient site-specific incorporation of multiple, different UAAs into a protein expressed in mammalian cells.

In one aspect, the invention provides a method of producing a protein comprising a first unnatural amino acid (UAA) and a second, different UAA (e.g., wherein the first and second UAA are each incorporated into the protein at specific, different positions in the protein polypeptide chain). The method comprises culturing a mammalian cell with: (i) a nucleic acid comprising a nucleotide sequence encoding a first tRNA comprising an anticodon that hybridizes to a first codon selected from UAG, UGA, and UAA, and is capable of being charged with the first UAA; (ii) a nucleic acid comprising a nucleotide sequence encoding a first aminoacyl-tRNA synthetase capable of charging the first tRNA with the first UAA; (iii) a nucleic acid comprising a nucleotide sequence encoding a second tRNA comprising an anticodon that hybridizes to a second codon selected from UAG, UGA, and UAA, and is capable of being charged with the second UAA, wherein the first and second tRNA do not contain the same anticodon; (iv) a nucleic acid comprising a nucleotide sequence encoding a second aminoacyl-tRNA synthetase capable of charging the second tRNA with the second UAA; and (v) a nucleic acid comprising a nucleotide sequence encoding the protein comprising the first codon and the second codon; under conditions that permit the first tRNA, when expressed in the cell and charged with the first UAA, to hybridize to the first codon and direct the incorporation of the first UAA into the protein, and the second tRNA, when expressed in the cell and charged with the second UAA, to hybridize to the second codon and direct the incorporation of the second UAA into the protein.

In certain embodiments, the expression yield of the protein comprising both the first and second UAA expressed by the cell is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the expression yield of a reference protein. For example, in certain embodiments, the amount of the protein comprising the first and second UAA expressed by the cell is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the amount of a reference protein expressed by the same cell or a similar cell. In certain embodiments, the reference protein is a protein that does not comprise the first and second UAA but is otherwise identical to the protein comprising the first and second UAA. For example, the reference protein may comprise a wild-type amino acid sequence, or comprise a wild-type amino acid residue at the positions corresponding to the first and second UAA.

In certain embodiments, the first or second tRNA is an analog or derivative of a prokaryotic tryptophanyl-tRNA, e.g., an E. coli tryptophanyl-tRNA. For example, the first or second tRNA may comprise a nucleotide sequence selected from any one of SEQ ID NOs: 49-54 or 108-113. In certain embodiments, the first or second aminoacyl-tRNA synthetase is an analog or derivative of a prokaryotic tryptophanyl-tRNA synthetase, e.g., an E. coli tryptophanyl-tRNA synthetase. For example, the first aminoacyl-tRNA synthetase may comprise an amino acid sequence selected from any one of SEQ ID NOs: 44-48. In certain embodiments, the first or second codon is UGA. In certain embodiments, the first or second UAA is a tryptophan analog, e.g., a non-naturally occurring tryptophan analog. In certain embodiments, the first or second UAA is 5-HTP or 5-AzW.

In certain embodiments, the first or second tRNA is an analog or derivative of a prokaryotic leucyl-tRNA, e.g., an E. coli leucyl-tRNA. For example, the first or second tRNA may comprise a nucleotide sequence selected from any one of SEQ ID NOs: 16-43. In certain embodiments, the first or second aminoacyl-tRNA synthetase is an analog or derivative of a prokaryotic leucyl-tRNA synthetase, e.g., an E. coli leucyl-tRNA synthetase. For example, the first aminoacyl-tRNA synthetase may comprise an amino acid sequence selected from any one of SEQ ID NOs: 1-15. In certain embodiments, the first or second codon is UAG. In certain embodiments, the first or second UAA is a leucine analog, e.g., a non-naturally occurring leucine analog. In certain embodiments, the first or second UAA is LCA or Cys-5-N3.

In certain embodiments, the first or second tRNA is an analog or derivative of a prokaryotic tyrosyl-tRNA, e.g., an E. coli tyrosyl-tRNA. For example, the first or second tRNA may comprise a nucleotide sequence selected from any one of SEQ ID NOs: 68-69 or 104-105. In certain embodiments, the first or second aminoacyl-tRNA synthetase is an analog or derivative of a prokaryotic tyrosyl-tRNA synthetase, e.g., an E. coli tyrosyl-tRNA synthetase. For example, the first aminoacyl-tRNA synthetase may comprise the amino acid sequence of SEQ ID NO: 70. In certain embodiments, the first or second codon is UAG. In certain embodiments, the first or second UAA is a tyrosine analog, e.g., a non-naturally occurring tyrosine analog. In certain embodiments, the first or second UAA is OmeY, AzF, or OpropY.

In certain embodiments, the first or second tRNA is an analog or derivative of an archael pyrrolysyl-tRNA, e.g., an M. barkeri pyrrolysyl-tRNA. For example, the first or second tRNA may comprise a nucleotide sequence selected from any one of SEQ ID NOs: 72-100 or 106-107. In certain embodiments, the first or second aminoacyl-tRNA synthetase is an analog or derivative of an archael pyrrolysyl-tRNA synthetase, e.g., an M. barkeri pyrrolysyl-tRNA synthetase. For example, the first aminoacyl-tRNA synthetase may comprise the amino acid sequence of SEQ ID NO: 101. In certain embodiments, the first or second codon is UAG. In certain embodiments, the first or second UAA is a pyrrolysine analog, e.g., a non-naturally occurring pyrrolysine analog. In certain embodiments, the first or second UAA is BocK, CpK, or AzK.

In certain embodiments, the protein is an antibody (or a fragment thereof, e.g., an antigen-binding fragment thereof), bispecific antibody, nanobody, affibody, viral protein, chemokine, cytokine, antigen, blood coagulation factor, hormone, growth factor, enzyme, cell signaling protein, or any other polypeptide or protein. In certain embodiments, the protein is an antibody (or a fragment thereof, e.g., an antigen-binding fragment thereof) and the first UAA and/or the second UAA are located in a constant region of the antibody.

In certain embodiments, the cell is a human cell, e.g., a human embryonic kidney (HEK) or a Chinese hamster ovary (CHO) cell.

In certain embodiments, the method further comprises contacting the cell with the first and/or second UAA. In certain embodiments, the method further comprises purifying the protein. In certain embodiments, the method further comprises chemically modifying the first and/or second UAA. For example, the chemical modification may comprise conjugation to a detectable label or molecule, e.g., a drug, e.g., a small molecule drug.

In another aspect, the invention provides a method of producing a protein comprising a tryptophan analog (e.g., a non-naturally occurring tryptophan analog) and a leucine analog (e.g., a non-naturally occurring leucine analog). The method comprises culturing a mammalian cell with: (i) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA comprising an anticodon that hybridizes to a UGA codon, and is capable of being charged with the tryptophan analog; (ii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA synthetase capable of charging the derivative of the E. coli tryptophanyl-tRNA with the tryptophan analog; (iii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli leucyl-tRNA comprising an anticodon that hybridizes to UAG codon, and is capable of being charged with the leucine analog; (iv) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli leucyl-tRNA synthetase capable of charging the derivative of the E. coli leucyl-tRNA with the leucine analog; and (v) a nucleic acid comprising a nucleotide sequence encoding the protein comprising the UGA codon and the UAG codon; under conditions that permit the derivative of the E. coli tryptophanyl-tRNA, when expressed in the cell and charged with the tryptophan analog, to hybridize to the UGA codon and direct the incorporation of the tryptophan into the protein, and the derivative of the E. coli leucyl-tRNA, when expressed in the cell and charged with the leucine analog, to hybridize to the UAG codon and direct the incorporation of the leucine analog into the protein.

In another aspect, the invention provides a method of producing a protein comprising a tryptophan analog (e.g., a non-naturally occurring tryptophan analog) and a tyrosine analog (e.g., a non-naturally occurring tyrosine analog). The method comprises culturing a mammalian cell with: (i) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA comprising an anticodon that hybridizes to a UGA codon, and is capable of being charged with the tryptophan analog; (ii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA synthetase capable of charging the derivative of the E. coli tryptophanyl-tRNA with the tryptophan analog; (iii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tyrosyl-tRNA comprising an anticodon that hybridizes to UAG codon, and is capable of being charged with the tyrosine analog; (iv) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tyrosyl-tRNA synthetase capable of charging the derivative of the E. coli tyrosyl-tRNA with the tyrosine analog; and (v) a nucleic acid comprising a nucleotide sequence encoding the protein comprising the UGA codon and the UAG codon; under conditions that permit the derivative of the E. coli tryptophanyl-tRNA, when expressed in the cell and charged with the tryptophan analog, to hybridize to the UGA codon and direct the incorporation of the tryptophan into the protein, and the derivative of the E. coli tyrosyl-tRNA, when expressed in the cell and charged with the tyrosine analog, to hybridize to the UAG codon and direct the incorporation of the tyrosine analog into the protein.

In another aspect, the invention provides a method of producing a protein comprising a tryptophan analog (e.g., a non-naturally occurring tryptophan analog) and a pyrrolysine analog (e.g., a non-naturally occurring pyrrolysine analog). The method comprises culturing a mammalian cell with: (i) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA comprising an anticodon that hybridizes to a UGA codon, and is capable of being charged with the tryptophan analog; (ii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA synthetase capable of charging the derivative of the E. coli tryptophanyl-tRNA with the tryptophan analog; (iii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an M. barkeri pyrrolysyl-tRNA comprising an anticodon that hybridizes to UAG codon, and is capable of being charged with the pyrrolysine analog; (iv) a nucleic acid comprising a nucleotide sequence encoding a derivative of an M. barkeri pyrrolysyl-tRNA synthetase capable of charging the derivative of the M. barkeri pyrrolysyl-tRNA with the pyrrolysine analog; and (v) a nucleic acid comprising a nucleotide sequence encoding the protein comprising the UGA codon and the UAG codon; under conditions that permit the derivative of the E. coli tryptophanyl-tRNA, when expressed in the cell and charged with the tryptophan analog, to hybridize to the UGA codon and direct the incorporation of the tryptophan into the protein, and the derivative of the E. coli pyrrolysyl-tRNA, when expressed in the cell and charged with the pyrrolysine analog, to hybridize to the UAG codon and direct the incorporation of the pyrrolysine analog into the protein.

In another aspect, the invention provides a protein comprising a first unnatural amino acid (UAA) and a second UAA produced by any of the foregoing methods.

In another aspect, the invention provides a protein expressed in a mammalian cell comprising a first unnatural amino acid (UAA) that is a tryptophan analog (e.g., a non-naturally occurring tryptophan analog) and a second UAA that is a leucine analog (e.g., a non-naturally occurring leucine analog). In certain embodiments, the tryptophan analog is selected from 5-HTP and 5-AzW and/or the leucine analog is selected from LCA and Cys-5-N3.

In another aspect, the invention provides a protein expressed in a mammalian cell comprising a first unnatural amino acid (UAA) that is a tryptophan analog (e.g., a non-naturally occurring tryptophan analog) and a second UAA that is a tyrosine analog (e.g., a non-naturally occurring tyrosine analog). In certain embodiments, the tryptophan analog is selected from 5-HTP and 5-AzW and/or the tyrosine analog is selected from OmeY, AzF, and OpropY UAA.

In another aspect, the invention provides a protein expressed in a mammalian cell comprising a first unnatural amino acid (UAA) that is a tryptophan analog (e.g., a non-naturally occurring tryptophan analog) and a second UAA that is a pyrrolysine analog (e.g., a non-naturally occurring pyrrolysine analog). In certain embodiments, the tryptophan analog is selected from 5-HTP and 5-AzW and/or the pyrrolysine analog is selected from BocK, CpK, AzK, and CpK.

In certain embodiments of any of the foregoing proteins, a detectable label or molecule (e.g., a drug, e.g., a small molecule drug) is covalently coupled to the protein via the first UAA and/or the second UAA. In certain embodiments, the protein is an antibody (or a fragment thereof, e.g., an antigen-binding fragment thereof), bispecific antibody, nanobody, affibody, viral protein, chemokine, cytokine, antigen, blood coagulation factor, hormone, growth factor, enzyme, cell signaling protein, or any other polypeptide or protein.

These and other aspects and features of the invention are described in the following detailed description and claims.

DESCRIPTION OF THE DRAWINGS

The invention can be more completely understood with reference to the following drawings.

FIG. 1 depicts a schematic overview of genetic code expansion using unnatural amino acids (UAAs).

FIGS. 2A-2C depicts a subset of UAAs that are exemplary substrates for a leucyl tRNA-synthetase.

FIG. 3 depicts a subset of UAAs that are exemplary substrates for a tryptophanyl tRNA-synthetase.

FIG. 4A depicts UAAs C5Az, LCA, and AzW. FIG. 4B depicts a synthetic route for C5Az. FIG. 4C depicts a synthetic route for AzW. FIGS. 4D-F depict synthetic routes for LCA.

FIG. 5 depicts a subset of UAAs that are exemplary substrates for a tyrosyl tRNA-synthetase.

FIG. 6 depicts a subset of UAAs that are exemplary substrates for a pyrrolysyl tRNA-synthetase.

FIG. 7 is a schematic overview of site-specific incorporation of two distinct unnatural amino acids (UAAs).

FIG. 8 depicts a comparison of TGA-stop codon suppression in mammalian cells of exemplary tRNA/aminoacyl tRNA synthetase (aaRS) pairs using a fluorescent reporter.

FIGS. 9A-9B demonstrate cross-reactivity of tested tRNA-synthetase pairs. FIG. 9A depicts a comparison of TAG-stop codon suppression in mammalian cells of LeuRS, EcTyrRS, PyIRS, and EcTrpRS pairs using a fluorescent reporter. FIG. 9B depicts a comparison of TGA-stop codon suppression in mammalian cells of LeuRS, EcTyrRS, PyIRS, and EcTrpRS pairs using a fluorescent reporter.

FIGS. 10A-10B demonstrate site-specific incorporation of two UAAs. FIG. 10A is a bar graph demonstrating site-specific incorporation efficiency of exemplary UAA pairs using EGFP fluorescence as a reporter for incorporation. FIG. 10B is an SDS-PAGE gel demonstrating production of exemplary proteins with two UAAs, using GFP protein production as a readout of site-specific incorporation.

FIGS. 11A-11J demonstrate electrospray-ionization mass spectrometry (ESI-MS) results of EGFP protein containing two UAAs. FIG. 11A is a histogram demonstrating mass spectrometry results of EGFP containing the exemplary UAAs HTP and BocK. FIG. 11B is a histogram demonstrating mass spectrometry results of EGFP containing the exemplary UAAs 5HTP and OMeY. FIG. 11C is a histogram demonstrating mass spectrometry results of EGFP containing the exemplary UAAs 5HTP and Cys-5-N3. FIG. 11D is a histogram demonstrating mass spectrometry results of EGFP containing the exemplary UAAs 5HTP and BocK. FIG. 11E is a histogram demonstrating mass spectrometry results of EGFP containing the exemplary UAAs 5HTP and Cyclopropene-K. FIG. 11F is a histogram demonstrating mass spectrometry results of EGFP containing the exemplary UAAs AzW and Cyclopropene-K. FIG. 11G is a histogram demonstrating mass spectrometry results of EGFP containing the exemplary UAAs 5HTP and AzK. FIG. 11H is a histogram demonstrating mass spectrometry results of fluorescent Diazo-labeled EGFP containing the exemplary UAAs 5HTP and AzK. FIG. 11I is a histogram demonstrating mass spectrometry results of DBCO-TAMRA-labeled EGFP containing the exemplary UAAs 5HTP and AzK. FIG. 11J is a histogram demonstrating mass spectrometry results of fluorescent Diazo and DBCO-TAMRA-labeled EGFP containing the exemplary UAAs 5HTP and AzK.

FIGS. 12A-12B are SDS-PAGE gels demonstrating production of fluorescently labeled EGFP with fluorescein (FIG. 12A) and TAMRA (FIG. 12B).

FIGS. 13A-13B demonstrate protein yield of IgG containing two UAAs. FIG. 13A is a bar graph demonstrating total protein yield in mg/L for IgG with UAA incorporation at T202 (heavy chain) and K113 (light chain). FIG. 13B is an SDS-PAGE gel of IgG with UAA incorporation at T202 and K113.

FIGS. 14A-14B demonstrate ESI-MS results of EGFP protein containing two UAAs. FIG. 14A is a histogram demonstrating mass spectrometry results of the PNGase-reduced IgG heavy chain (HC) containing the exemplary UAA HTP. FIG. 14B is a histogram demonstrating mass spectrometry results of PNGase-reduced IgG-light chain (LC) containing the exemplary UAA LCA.

FIG. 15 depicts the conserved regions of the IgG protein structure utilized for site-specific incorporation of multiple UAAs.

FIG. 16 depicts a schematic diagram of an antibody including the UAAs HTP and LCA, labeled with Click-Chemistry and Diazo coupling, e.g., for mass spectrometry studies.

FIGS. 17A-17E display high performance liquid chromatography hydrophobic interaction chromatography (HPLC-HIC) traces of an antibody including the UAAs HTP and LCA, labeled with Click-Chemistry and Diazo coupling.

DETAILED DESCRIPTION

The invention is based, in part, on the discovery of combinations of nonsense codons, tRNAs, aminoacyl-tRNA synthetases, and UAAs that allow for efficient site-specific incorporation of multiple, different UAAs into a protein produced in mammalian cells. Selection of the components that allow for successful, efficient incorporation of multiple UAAs in a protein is challenging given that there are over two million possible combinations of distinct aaRS, tRNA, UAA, codon, and pair combinations that could be used for such multisite incorporation. As a result, it is challenging to find specific combinations of these elements that are suitable for use for site-specific incorporation of two distinct bioconjugation handles.

In one aspect, the invention provides a method of producing a protein comprising a first unnatural amino acid (UAA) and a second, different UAA (e.g., wherein the first and second UAA are each incorporated into the protein at specific, different positions in the protein polypeptide chain). The method comprises culturing a mammalian cell with: (i) a nucleic acid comprising a nucleotide sequence encoding a first tRNA comprising an anticodon that hybridizes to a first codon selected from UAG, UGA, and UAA, and is capable of being charged with the first UAA; (ii) a nucleic acid comprising a nucleotide sequence encoding a first aminoacyl-tRNA synthetase capable of charging the first tRNA with the first UAA; (iii) a nucleic acid comprising a nucleotide sequence encoding a second tRNA comprising an anticodon that hybridizes to a second codon selected from UAG, UGA, and UAA, and is capable of being charged with the second UAA, wherein the first and second tRNA do not contain the same anticodon; (iv) a nucleic acid comprising a nucleotide sequence encoding a second aminoacyl-tRNA synthetase capable of charging the second tRNA with the second UAA; and (v) a nucleic acid comprising a nucleotide sequence encoding the protein comprising the first codon and the second codon; under conditions that permit the first tRNA, when expressed in the cell and charged with the first UAA, to hybridize to the first codon and direct the incorporation of the first UAA into the protein, and the second tRNA, when expressed in the cell and charged with the second UAA, to hybridize to the second codon and direct the incorporation of the second UAA into the protein.

In certain embodiments, the expression yield of the protein comprising the first and second UAA expressed by the cell is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the expression yield of a reference protein. For example, in certain embodiments, the amount of protein comprising the first and second UAA expressed by the cell is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the amount of a reference protein expressed by the same cell or a similar cell. In certain embodiments, the reference protein is a protein that does not comprise the first and second UAA but is otherwise identical to the protein comprising the first and second UAA. For example, the reference protein may comprise a wild-type amino acid sequence, or comprise a wild-type amino acid residue at the positions corresponding to the first and second UAA. Protein expression may be measured by any method known in the art, including for example, Western blot or ELISA. Expression may be measured by measuring protein concentration (e.g., by ultraviolet (UV) absorption at 280 nm or Bradford assay) in a solution of defined volume and purity following purification of the protein. Expression of a fluorescent protein (e.g., a reporter protein) may be measured by fluorescence microscopy as described in Examples 2-4 herein. In certain embodiments, a disclosed method will result in an expression yield of an EGFP reporter protein containing a first and second UAA (e.g., an EGFP reporter protein encoded by a nucleotide sequence containing two stop codons, e.g., EGFP-39TAG-151TGA containing stop codons TAG and TGA, as described in Example 4) that is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the expression yield of a corresponding EGFP reporter protein that does not contain the first and second UAA (e.g., a wild-type EGFP protein).

In another aspect, the invention provides a method of producing a protein comprising a tryptophan analog and a leucine analog. The method comprises culturing a mammalian cell with: (i) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA comprising an anticodon that hybridizes to a UGA codon, and is capable of being charged with the tryptophan analog; (ii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA synthetase capable of charging the derivative of the E. coli tryptophanyl-tRNA with the tryptophan analog; (iii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli leucyl-tRNA comprising an anticodon that hybridizes to UAG codon, and is capable of being charged with the leucine analog; (iv) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli leucyl-tRNA synthetase capable of charging the derivative of the E. coli leucyl-tRNA with the leucine analog; and (v) a nucleic acid comprising a nucleotide sequence encoding the protein comprising the UGA codon and the UAG codon; under conditions that permit the derivative of the E. coli tryptophanyl-tRNA, when expressed in the cell and charged with the tryptophan analog, to hybridize to the UGA codon and direct the incorporation of the tryptophan into the protein, and the derivative of the E. coli leucyl-tRNA, when expressed in the cell and charged with the leucine analog, to hybridize to the UAG codon and direct the incorporation of the leucine analog into the protein.

In another aspect, the invention provides a method of producing a protein comprising a tryptophan analog and a tyrosine analog. The method comprises culturing a mammalian cell with: (i) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA comprising an anticodon that hybridizes to a UGA codon, and is capable of being charged with the tryptophan analog; (ii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA synthetase capable of charging the derivative of the E. coli tryptophanyl-tRNA with the tryptophan analog; (iii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tyrosyl-tRNA comprising an anticodon that hybridizes to UAG codon, and is capable of being charged with the tyrosine analog; (iv) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tyrosyl-tRNA synthetase capable of charging the derivative of the E. coli tyrosyl-tRNA with the tyrosine analog; and (v) a nucleic acid comprising a nucleotide sequence encoding the protein comprising the UGA codon and the UAG codon; under conditions that permit the derivative of the E. coli tryptophanyl-tRNA, when expressed in the cell and charged with the tryptophan analog, to hybridize to the UGA codon and direct the incorporation of the tryptophan into the protein, and the derivative of the E. coli tyrosyl-tRNA, when expressed in the cell and charged with the tyrosine analog, to hybridize to the UAG codon and direct the incorporation of the tyrosine analog into the protein.

In another aspect, the invention provides a method of producing a protein comprising a tryptophan analog and a pyrrolysine analog. The method comprises culturing a mammalian cell with: (i) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA comprising an anticodon that hybridizes to a UGA codon, and is capable of being charged with the tryptophan analog; (ii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA synthetase capable of charging the derivative of the E. coli tryptophanyl-tRNA with the tryptophan analog; (iii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an M. barkeri pyrrolysyl-tRNA comprising an anticodon that hybridizes to UAG codon, and is capable of being charged with the pyrrolysine analog; (iv) a nucleic acid comprising a nucleotide sequence encoding a derivative of an M. barkeri pyrrolysyl-tRNA synthetase capable of charging the derivative of the M. barkeri pyrrolysyl-tRNA with the pyrrolysine analog; and (v) a nucleic acid comprising a nucleotide sequence encoding the protein comprising the UGA codon and the UAG codon; under conditions that permit the derivative of the E. coli tryptophanyl-tRNA, when expressed in the cell and charged with the tryptophan analog, to hybridize to the UGA codon and direct the incorporation of the tryptophan into the protein, and the derivative of the E. coli pyrrolysyl-tRNA, when expressed in the cell and charged with the pyrrolysine analog, to hybridize to the UAG codon and direct the incorporation of the pyrrolysine analog into the protein.

In another aspect, the invention provides a protein comprising a first unnatural amino acid (UAA) and a second UAA made by any of the foregoing methods.

In another aspect, the invention provides a protein expressed in a mammalian cell comprising a first unnatural amino acid (UAA) that is a tryptophan analog and a second UAA that is a leucine analog. In certain embodiments, the tryptophan analog is selected from 5-HTP and 5-AzW and/or the leucine analog is selected from LCA and Cys-5-N3.

In another aspect, the invention provides a protein expressed in a mammalian cell comprising a first unnatural amino acid (UAA) that is a tryptophan analog and a second UAA that is a tyrosine analog. In certain embodiments, the tryptophan analog is selected from 5-HTP and 5-AzW and/or the tyrosine analog is selected from OmeY, AzF, and OpropY UAA.

In another aspect, the invention provides a protein expressed in a mammalian cell comprising a first unnatural amino acid (UAA) that is a tryptophan analog and a second UAA that is a pyrrolysine analog. In certain embodiments, the tryptophan analog is selected from 5-HTP and 5-AzW and/or the pyrrolysine analog is selected from BocK, CpK, AzK, and CpK.

In another aspect, the invention provides a composition comprising any of the foregoing proteins. In certain embodiments, the purity of the comprising the first and second UAA in composition is at least 50% (i.e., the protein comprising the first and second UAA is at least 50% of the total protein in the composition). In certain embodiments, the purity of the protein in composition is at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%. In certain embodiments, less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, less than 5%, less than 4%, less than 3%, less than 2% or less than 1% of the total protein in the composition is any protein that is otherwise identical to the protein comprising the first and second UAA but does not comprise the first or second UAA.

As used herein, the term “orthogonal” refers to a molecule (e.g., an orthogonal tRNA or an orthogonal aminoacyl-tRNA synthetase) that is used with reduced efficiency by an expression system of interest (e.g., an endogenous cellular translation system). For example, an orthogonal tRNA in a translation system of interest is aminoacylated by any endogenous aminoacyl-tRNA synthetase of the translation system of interest with reduced or even zero efficiency, when compared to aminoacylation of an endogenous tRNA by an endogenous aminoacyl-tRNA synthetase. In another example, an orthogonal aminoacyl-tRNA synthetase aminoacylates any endogenous tRNA in the translation system of interest with reduced or even zero efficiency, as compared to aminoacylation of an endogenous tRNA by an endogenous aminoacyl-tRNA synthetase.

Various features and aspects of the invention are discussed in more detail below.

I. Aminoacyl-tRNA Synthetases

The invention relates to engineered aminoacyl-tRNA synthetases (or aaRSs) capable of charging a tRNA with an unnatural amino acid for incorporation into a protein. As used herein, the term “aminoacyl-tRNA synthetase” refers to any enzyme, or a functional fragment thereof, that charges, or is capable of charging, a tRNA with an amino acid (e.g., an unnatural amino acid) for incorporation into a protein. As used herein, the term “functional fragment” of an aminoacyl-tRNA synthetase refers to fragment of a full-length aminoacyl-tRNA synthetase that retains, for example, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the enzymatic activity of the corresponding full-length tRNA synthetase (e.g., a naturally occurring tRNA synthetase). Aminoacyl-tRNA synthetase enzymatic activity may be assayed by any method known in the art. For example, in vitro aminoacylation assays are described in Hoben et al. (1985) METHODS ENZYMOL. 113:55-59 and in U.S. Patent Application Publication No. 2003/0228593 and cell-based aminoacylation assays are described in U.S. Patent Application Publication Nos. 2003/0082575 and 2005/0009049. In certain embodiments, the functional fragment comprises at least 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, or 800 consecutive amino acids present in a full-length tRNA synthetase (e.g., a naturally occurring aminoacyl-tRNA synthetase).

The term aminoacyl-tRNA synthetase includes variants (i.e., muteins) having one or more mutations (e.g., amino acid substitutions, deletions, or insertions) relative to a wild-type aminoacyl-tRNA synthetase sequence. In certain embodiments, an aminoacyl-tRNA synthetase mutein may comprise, consist, or consist essentially of, a single mutation (e.g., a mutation contemplated herein), or a combination of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more than 15 mutations (e.g., mutations contemplated herein). It is contemplated that an aminoacyl-tRNA synthetase mutein may comprise, consist, or consist essentially 1-15, 1-10, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 2-15, 2-10, 2-7, 2-6, 2-5, 2-4, 2-3, 3-15, 3-10, 3-7, 3-6, 3-5, or 4-10, 4-7, 4-6, 4-5, 5-10, 5-7, 5-6, 6-10, 6-7, 7-10, 7-8, or 8-10 mutations (e.g., mutations contemplated herein).

An aminoacyl-tRNA synthetase mutein may comprise a conservative substitution relative to a wild-type sequence or a sequence disclosed herein. As used herein, the term “conservative substitution” refers to a substitution with a structurally similar amino acid. For example, conservative substitutions may include those within the following groups: Ser and Cys; Leu, Ile, and Val; Glu and Asp; Lys and Arg; Phe, Tyr, and Trp; and Gln, Asn, Glu, Asp, and His. Conservative substitutions may also be defined by the BLAST (Basic Local Alignment Search Tool) algorithm, the BLOSUM substitution matrix (e.g., BLOSUM 62 matrix), or the PAM substitution:p matrix (e.g., the PAM 250 matrix).

In certain embodiments, the substrate specificity of the aminoacyl-tRNA synthetase mutein is altered relative to a corresponding (or template) wild-type aminoacyl-tRNA synthetase such that only a desired unnatural amino acid, but not any of the common 20 amino acids, is charged to the substrate tRNA.

An aminoacyl-tRNA synthetase may be derived from a bacterial source, e.g., Escherichia coli, Thermus thermophilus, or Bacillus stearothermphilus. An aminoacyl-tRNA synthetase may also be derived from an archaeal source, e.g, from the Methanosarcinacaea or Desulfitobacterium families, any of the M. barkeri (Mb), M. alvus (Ma), M. mazei (Mm) or D. hafnisense (Dh) families, Methanobacterium thermoautotrophicum, Haloferax volcanii, Halobacterium species NRC-1, or Archaeoglobus fulgidus. In other embodiments, eukaryotic sources can also be used, for example, plants, algae, protists, fungi, yeasts, or animals (e.g., mammals, insects, arthropods, etc.). As used herein, the terms “derivative” or “derived from” refer to a component that is isolated from or made using information from a specified molecule or organism. As used herein, the term “analog” refers to a component (e.g., a tRNA, tRNA synthetase, or unnatural amino acid) that is derived from or analogous with (in terms of structure and/or function) a reference component (e.g., a wild-type tRNA, a wild-type tRNA synthetase, or a natural amino acid). In certain embodiments, derivatives or analogs have at least 40%, 50%, 60%, 70%, 80%, 90%, 100% or more of a given activity as a reference or originator component (e.g., wild type component).

It is contemplated that the aminoacyl-tRNA synthetase may aminoacylate a substrate tRNA in vitro or in vivo, and can be provided to a translation system (e.g., an in vitro translation system or a cell) as a polypeptide or protein, or as a polynucleotide that encodes the aminoacyl-tRNA synthetase.

In certain embodiments, the aminoacyl-tRNA synthetase is derived from an E. coli leucyl-tRNA synthetase and, for example, the aminoacyl-tRNA synthetase preferentially aminoacylates an E. coli leucyl tRNA (or a variant thereof) with a leucine analog over the naturally-occurring leucine amino acid.

For example, the aminoacyl-tRNA synthetase may comprise SEQ ID NO: 1, or an amino acid sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 1. In certain embodiments, the aminoacyl-tRNA synthetase comprises SEQ ID NO: 1, or a functional fragment or variant thereof, and with one, two, three, four, five or more of the following mutations: (i) a substitution of a glutamine residue at a position corresponding to position 2 of SEQ ID NO: 1, e.g., a substitution by glutamic acid (Q2E); (ii) a substitution of a glutamic acid residue at a position corresponding to position 20 of SEQ ID NO: 1, e.g., a substitution by lysine (E20K), methionine (E20M), or valine (E20V); (iii) a substitution of a methionine residue at a position corresponding to position 40 of SEQ ID NO: 1, e.g., a substitution by isoleucine (M40I) or valine (M40V); (iv) a substitution of a leucine residue at a position corresponding to position 41 of SEQ ID NO: 1, e.g., a substitution by serine (L41S), valine (L41V), or alanine (L41A); (v) a substitution of a threonine residue at a position corresponding to position 252 of SEQ ID NO: 1, e.g., a substitution by alanine (T252A) or arginine (T252R); (vi) a substitution of a tyrosine residue at a position corresponding to position 499 of SEQ ID NO: 1, e.g., a substitution by isoleucine (Y499I), serine (Y499S), alanine (Y499A), or histidine (Y499H); (vii) a substitution of a tyrosine residue at a position corresponding to position 527 of SEQ ID NO: 1, e.g., a substitution by alanine (Y527A), leucine (Y527L), isoleucine (Y527I), valine (Y527V), or glycine (Y527G); or (viii) a substitution of a histidine residue at a position corresponding to position 537 of SEQ ID NO: 1, e.g., a substitution by glycine (H537G), or any combination of the foregoing.

In certain embodiments, the aminoacyl-tRNA synthetase comprises (i) at least one substitution (e.g., a substitution with a hydrophobic amino acid) at a position corresponding to His537 of SEQ ID NO: 1, (ii) at least one amino acid substitution selected from E20V, E20M, L41V, L41A, Y499H, Y499A, Y527I, Y527V, Y527G, and any combination thereof, (iii) at least one amino acid substitution selected from E20K and L41S and any combination thereof and at least one amino acid substitution selected from M40I, T252A, Y499I, and Y527A, and any combination thereof, or (iv) a combination of two or more of (i), (ii) and (iii), for example, (i) and (ii), (i) and (iii), (ii) and (iii) and (i), (ii) and (iii).

In certain embodiments, the aminoacyl-tRNA synthetase comprises a substitution of a glutamic acid residue at a position corresponding to position 20 of SEQ ID NO: 1, e.g., a substitution with an amino acid other than a Glu or Lys, e.g., a substitution with a hydrophobic amino acid (e.g., Leu, Val, or Met). In certain embodiments, the aminoacyl-tRNA synthetase comprises a substitution of a leucine residue at a position corresponding to position 41 of SEQ ID NO: 1, e.g., a substitution with an amino acid other than a Leu or Ser, e.g., a substitution with a hydrophobic amino acid other than Leu (e.g., Gly, Ala, Val, or Met). In certain embodiments, the aminoacyl-tRNA synthetase comprises a substitution of a tyrosine residue at a position corresponding to position 499 of SEQ ID NO: 1, e.g., a substitution with a small hydrophobic amino acid (e.g., Gly, Ala, or Val) or a substitution with a positively charged amino acid (e.g., Lys, Arg, or His). In certain embodiments, the aminoacyl-tRNA synthetase comprises a substitution of a tyrosine residue at a position corresponding to position 527 of SEQ ID NO: 1, e.g., a substitution with a hydrophobic amino acid other than Ala or Leu (e.g., Gly, Ile, Met, or Val). In certain embodiments, the tRNA synthetase mutein comprises L41V.

In certain embodiments, the aminoacyl-tRNA synthetase comprises a combination of mutations selected from: (i) Q2E, E20K, M40I, L41S, T252A, Y499I, Y527A, and H537G; (ii) Q2E, E20K, M40V, L41S, T252R, Y499S, Y527L, and H537G; (iii) Q2E, M40I, T252A, Y499I, Y527A, and H537G; (iv) Q2E, E20M, M40I, L41S, T252A, Y499I, Y527A, and H537G; (v) Q2E, E20V, M40I, L41S, T252A, Y499I, Y527A, and H537G; (vi) Q2E, E20K, M40I, L41V, T252A, Y499I, Y527A, and H537G; (vii) Q2E, E20K, M40I, L41A, T252A, Y499I, Y527A, and H537G; (viii) Q2E, E20K, M40I, L41S, T252A, Y499A, Y527A, and H537G; (ix) Q2E, E20K, M40I, L41S, T252A, Y499H, Y527A, and H537G; (x) Q2E, E20K, M40I, L41S, T252A, Y499I, Y527I, and H537G; (xi) Q2E, E20K, M40I, L41S, T252A, Y499I, Y527V, and H537G; (xii) Q2E, E20K, M40I, L41S, T252A, Y499I, Y527G, and H537G; (xiii) E20K, M40I, L41S, T252A, Y499I, Y527A, and H537G; (xiv) E20M, M40I, L41S, T252A, Y499I, Y527A, and H537G; (xv) E20V, M40I, L41S, T252A, Y499I, Y527A, and H537G; (xvi) E20K, M40I, L41V, T252A, Y499I, Y527A, and H537G; (vii) E20K, M40I, L41A, T252A, Y499L, Y527A, and H537G; (xviii) E20K, M40I, L41S, T252A, Y499A, Y527A, and H537G; (xix) E20K, M40I, L41S, T252A, Y499H, Y527A, and H537G; (xx) E20K, M40I, L41S, T252A, Y499I, Y527I, and H537G; (xxi) E20K, M40I, L41S, T252A, Y499I, Y527V, and H537G; and (xxii) E20K, M40I, L41S, T252A, Y499L, Y527G, and H537G.

In certain embodiments, the aminoacyl-tRNA synthetase comprises the amino acid sequence of any one of SEQ ID NOs: 2-13, or an amino acid sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOs: 2-13.

In certain embodiments, the tRNA synthetase mutein comprises the amino acid sequence of SEQ ID NO: 14, wherein X2 is Q or E, X20 is E, K, V or M, X40 is M, I, or V, X41 is L, S, V, or A, X252 is T, A, or R, X499 is Y, A, I, H, or S, X527 is Y, A, I, L, or V, and X537 is H or G, and the tRNA synthetase mutein comprises at least one mutation (for example, 2, 3, 4, 5, 6, 7, 8, 9, or more mutations) relative to SEQ ID NO: 1. In certain embodiments, the tRNA synthetase mutein comprises the amino acid sequence of SEQ ID NO: 15, wherein X20 is K, V or M, X41 is S, V, or A, X499 is A, I, or H, and X527 is A, I, or V, and the tRNA synthetase mutein comprises at least one mutation relative to SEQ ID NO: 1.

In certain embodiments, the aminoacyl-tRNA synthetase is derived from an E. coli tryptophanyl-tRNA synthetase and, for example, the aminoacyl-tRNA synthetase preferentially aminoacylates an E. coli tryptophanyl tRNA (or a variant thereof) with a tryptophan analog over the naturally-occurring tryptophan amino acid.

For example, the aminoacyl-tRNA synthetase may comprise SEQ ID NO: 44, or an amino acid sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 44. In certain embodiments, the aminoacyl-tRNA synthetase comprises SEQ ID NO: 44, or a functional fragment or variant thereof, but with one or more of the following mutations: (i) a substitution of a serine residue at a position corresponding to position 8 of SEQ ID NO: 44, e.g., a substitution by alanine (S8A); (ii) a substitution of a valine residue at a position corresponding to position 144 of SEQ ID NO: 44, e.g., a substitution by serine (V144S), glycine (V144G) or alanine (V144A); (iii) a substitution of a valine residue at a position corresponding to position 146 of SEQ ID NO: 44, e.g., a substitution by alanine (V146A), isoleucine (V146I), or cysteine (V146C). In certain embodiments, the aminoacyl-tRNA synthetase comprises a combination of mutations selected from: (i) S8A, V144S, and V146A, (ii) S8A, V144G, and V146I, (iii) S8A, V144A, and V146A, and (iv) S8A, V144G, and V146C.

In certain embodiments, the aminoacyl-tRNA synthetase comprises the amino acid sequence of any one of SEQ ID NOs: 45-48, or an amino acid sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOs: 45-48.

In certain embodiments, the aminoacyl-tRNA synthetase is derived from an E. coli tyrosyl-tRNA synthetase and, for example, the aminoacyl-tRNA synthetase preferentially aminoacylates an E. coli tyrosyl tRNA (or a variant thereof) with a tyrosine analog over the naturally-occurring tryptophan amino acid. For example, the aminoacyl-tRNA synthetase may comprise SEQ ID NO: 70, or an amino acid sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 70, or a functional fragment or variant thereof.

In certain embodiments, the aminoacyl-tRNA synthetase is derived from an M. barkeri pyrrolysyl-tRNA synthetase and, for example, the aminoacyl-tRNA synthetase preferentially aminoacylates an M. barkeri pyrrolysyl tRNA (or a variant thereof) with a pyrrolysine analog over the naturally-occurring pyrrolysine amino acid. For example, the aminoacyl-tRNA synthetase may comprise SEQ ID NO: 101, or an amino acid sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 101, or a functional fragment or variant thereof.

Sequence identity may be determined in various ways that are within the skill of a person skilled in the art, e.g., using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. BLAST (Basic Local Alignment Search Tool) analysis using the algorithm employed by the programs blastp, blastn, blastx, tblastn and tblastx (Karlin et al., (1990) PROC. NATL. ACAD. SCI. USA 87:2264-2268; Altschul, (1993) J. MOL. EVOL. 36:290-300; Altschul et al., (1997) NUCLEIC ACIDS RES. 25:3389-3402, incorporated by reference herein) are tailored for sequence similarity searching. For a discussion of basic issues in searching sequence databases see Altschul et al., (1994) NATURE GENETICS 6:119-129, which is fully incorporated by reference herein. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. The search parameters for histogram, descriptions, alignments, expect (i.e., the statistical significance threshold for reporting matches against database sequences), cutoff, matrix and filter are at the default settings. The default scoring matrix used by blastp, blastx, tblastn, and tblastx is the BLOSUM62 matrix (Henikoff et al., (1992) PROC. NATL. ACAD. SCI. USA 89:10915-10919, fully incorporated by reference herein). Four blastn parameters may be adjusted as follows: Q=10 (gap creation penalty); R=10 (gap extension penalty); wink=1 (generates word hits at every wink.sup.th position along the query); and gapw=16 (sets the window width within which gapped alignments are generated). The equivalent blastp parameter settings may be Q=9; R=2; wink=1; and gapw=32. Searches may also be conducted using the NCBI (National Center for Biotechnology Information) BLAST Advanced Option parameter (e.g.: -G, Cost to open gap [Integer]: default=5 for nucleotides/11 for proteins; -E, Cost to extend gap [Integer]: default=2 for nucleotides/1 for proteins; -q, Penalty for nucleotide mismatch [Integer]: default=−3; -r, reward for nucleotide match [Integer]: default=1; -e, expect value [Real]: default=10; —W, wordsize [Integer]: default=11 for nucleotides/28 for megablast/3 for proteins; -y, Dropoff (X) for blast extensions in bits: default=20 for blastn/7 for others; -X, X dropoff value for gapped alignment (in bits): default=15 for all programs, not applicable to blastn; and -Z, final X dropoff value for gapped alignment (in bits): 50 for blastn, 25 for others). ClustalW for pairwise protein alignments may also be used (default parameters may include, e.g., Blosum62 matrix and Gap Opening Penalty=10 and Gap Extension Penalty=0.1). A Bestfit comparison between sequences, available in the GCG package version 10.0, uses DNA parameters GAP=50 (gap creation penalty) and LEN=3 (gap extension penalty). The equivalent settings in Bestfit protein comparisons are GAP=8 and LEN=2.

Methods for producing proteins, e.g., aminoacyl-tRNA synthetases, are known in the art. For example, DNA molecules encoding a protein of interest can be synthesized chemically or by recombinant DNA methodologies. The resulting DNA molecules encoding the protein interest can be ligated to other appropriate nucleotide sequences, including, for example, expression control sequences, to produce conventional gene expression constructs (i.e., expression vectors) encoding the desired protein. Production of defined gene constructs is within routine skill in the art.

Nucleic acids encoding desired proteins (e.g, aminoacyl-tRNA synthetases) can be incorporated (ligated) into expression vectors, which can be introduced into host cells through conventional transfection or transformation techniques. Exemplary host cells are E. coli cells, Chinese hamster ovary (CHO) cells, human embryonic kidney 293 (HEK 293) cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), and myeloma cells. Transformed host cells can be grown under conditions that permit the host cells to express the desired protein.

Specific expression and purification conditions will vary depending upon the expression system employed. For example, if a gene is to be expressed in E. coli, it is first cloned into an expression vector by positioning the engineered gene downstream from a suitable bacterial promoter, e.g., Trp or Tac, and a prokaryotic signal sequence. The expressed protein may be secreted. The expressed protein may accumulate in refractile or inclusion bodies, which can be harvested after disruption of the cells by French press or sonication. The refractile bodies then are solubilized, and the protein may be refolded and/or cleaved by methods known in the art.

If the engineered gene is to be expressed in eukaryotic host cells, e.g., CHO cells, it is first inserted into an expression vector containing a suitable eukaryotic promoter, a secretion signal, a poly A sequence, and a stop codon. Optionally, the vector or gene construct may contain enhancers and introns. The gene construct can be introduced into eukaryotic host cells using conventional techniques.

A protein of interest (e.g, an aminoacyl-tRNA synthetase) can be produced by growing (culturing) a host cell transfected with an expression vector encoding such a protein under conditions that permit expression of the protein. Following expression, the protein can be harvested and purified or isolated using techniques known in the art, e.g., affinity tags such as glutathione-S-transferase (GST) or histidine tags.

Additional methods for producing aminoacyl-tRNA synthetases, and for altering the substrate specificity of the synthetase can be found in U.S. Patent Application Publication Nos. 2003/0108885 and 2005/0009049, Hamano-Takaku et al. (2000) JOURNAL OF BIOL. CHEM. 275(51):40324-40328, Kiga et al. (2002) PROC. NATL. ACAD. SCI. USA 99(15): 9715-9723, and Francklyn et al. (2002) RNA, 8:1363-1372.

The invention also encompasses nucleic acids encoding aminoacyl-tRNA synthetases disclosed herein. For example, nucleotide sequences encoding leucyl-tRNA synthetase muteins disclosed herein are depicted in SEQ ID NOs: 55-67. Accordingly, the invention provides a nucleic acid comprising the nucleotide sequence of any one of SEQ ID NOs: 55-67, or a nucleotide sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOs: 55-67. The invention also provides a nucleic acid comprising a nucleotide sequence encoding the amino acid sequence encoded by any one of SEQ ID NOs: 55-67, or a nucleotide sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to a nucleotide sequence encoding the amino acid sequence encoded by any one of SEQ ID NOs: 55-67.

A nucleotide sequence encoding a tryptophanyl-tRNA synthetase disclosed herein is depicted in SEQ ID NO: 103. Accordingly, the invention provides a nucleic acid comprising the nucleotide sequence of SEQ ID NO: 103, or a nucleotide sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 103. The invention also provides a nucleic acid comprising a nucleotide sequence encoding the amino acid sequence encoded by SEQ ID NO: 103, or a nucleotide sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to a nucleotide sequence encoding the amino acid sequence encoded by SEQ ID NO: 103.

A nucleotide sequence encoding a tyrosyl-tRNA synthetase disclosed herein is depicted in SEQ ID NO: 71. Accordingly, the invention provides a nucleic acid comprising the nucleotide sequence of SEQ ID NO: 71, or a nucleotide sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 71. The invention also provides a nucleic acid comprising a nucleotide sequence encoding the amino acid sequence encoded by SEQ ID NO: 71, or a nucleotide sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to a nucleotide sequence encoding the amino acid sequence encoded by SEQ ID NO: 71.

A nucleotide sequence encoding a pyrrolysyl-tRNA synthetase disclosed herein is depicted in SEQ ID NO: 102. Accordingly, the invention provides a nucleic acid comprising the nucleotide sequence of SEQ ID NO: 102, or a nucleotide sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 102. The invention also provides a nucleic acid comprising a nucleotide sequence encoding the amino acid sequence encoded by SEQ ID NO: 102, or a nucleotide sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to a nucleotide sequence encoding the amino acid sequence encoded by SEQ ID NO: 102.

II. tRNAs

The invention relates to transfer RNAs (tRNAs) that mediate the incorporation of unnatural amino acids into proteins.

During protein synthesis, a tRNA molecule delivers an amino acid to a ribosome for incorporation into a growing protein (polypeptide) chain. tRNAs typically are about 70 to 100 nucleotides in length. Active tRNAs contain a 3′ CCA sequence that may be transcribed into the tRNA during its synthesis or may be added later during post-transcriptional processing. During aminoacylation, the amino acid that is attached to a given tRNA molecule is covalently attached to the 2′ or 3′ hydroxyl group of the 3′-terminal ribose to form an aminoacyl-tRNA (aa-tRNA). It is understood that an amino acid can spontaneously migrate from the 2′-hydroxyl group to the 3′-hydroxyl group and vice versa, but it is incorporated into a growing protein chain at the ribosome from the 3-OH position. A loop at the other end of the folded aa-tRNA molecule contains a sequence of three bases known as the anticodon. When this anticodon sequence hybridizes or base-pairs with a complementary three-base codon sequence in a ribosome-bound mRNA, the aa-tRNA binds to the ribosome and its amino acid is incorporated into the polypeptide chain being synthesized by the ribosome. Because all tRNAs that base-pair with a specific codon are aminoacylated with a single specific amino acid, the translation of the genetic code is effected by tRNAs. Each of the 61 non-termination codons in an mRNA directs the binding of its cognate aa-tRNA and the addition of a single specific amino acid to the growing polypeptide chain being synthesized by the ribosome. The term “cognate” refers to components that function together, e.g., a tRNA and an aminoacyl-tRNA synthetase.

Suppressor tRNAs are modified tRNAs that alter the reading of a mRNA in a given translation system. For example, a suppressor tRNA may read through a codon such as a stop codon, a four base codon, or a rare codon. The use of the word in suppressor is based on the fact, that under certain circumstance, the modified tRNA “suppresses” the typical phenotypic effect of the codon in the mRNA. Suppressor tRNAs typically contain a mutation (modification) in either the anticodon, changing codon specificity, or at some position that alters the aminoacylation identity of the tRNA. The term “suppression activity” refers to the ability of a tRNA, e.g., a suppressor tRNA, to read through a codon (e.g., a premature stop codon) that would not be read through by the endogenous translation machinery in a system of interest.

In certain embodiments, a tRNA (e.g., a suppressor tRNA) contains a modified anticodon region, such that the modified anticodon hybridizes with a different codon than the corresponding naturally occurring anticodon.

In certain embodiments, a tRNA comprises an anticodon that hybridizes to a codon selected from UAG (i.e., an “amber” termination codon), UGA (i.e., an “opal” termination codon), and UAA (i.e., an “ochre” termination codon).

In certain embodiments, a tRNA comprises an anticodon that hybridizes to a non-standard codon, e.g., a 4- or 5-nucleotide codon. Examples of four base codons include AGGA, CUAG, UAGA, and CCCU. Examples of five base codons include AGGAC, CCCCU, CCCUC, CUAGA, CUACU, and UAGGC. tRNAs comprising an anticodon that hybridizes to a non-standard codon, e.g., a 4- or 5-nucleotide codon, and methods of using such tRNAs to incorporate unnatural amino acids into proteins are described, for example, in Moore et al. (2000) J. MOL. BIOL. 298:195; Hohsaka et al. (1999) J. AM. CHEM. SOC. 121:12194; Anderson et al. (2002) CHEMISTRY AND BIOLOGY 9:237-244; Magliery (2001) J. MoL. BIOL. 307: 755-769; and PCT Publication No. WO2005/007870.

As used herein, the term “tRNA” includes variants having one or more mutations (e.g., nucleotide substitutions, deletions, or insertions) relative to a reference (e.g., a wild-type) tRNA sequence. In certain embodiments, a tRNA may comprise, consist, or consist essentially of, a single mutation (e.g., a mutation contemplated herein), or a combination of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more than 15 mutations (e.g., mutations contemplated herein). It is contemplated that a tRNA may comprise, consist, or consist essentially 1-15, 1-10, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 2-15, 2-10, 2-7, 2-6, 2-5, 2-4, 2-3, 3-15, 3-10, 3-7, 3-6, 3-5, or 3-4 mutations (e.g., mutations contemplated herein).

In certain embodiments, a variant suppressor tRNA has increased activity to incorporate an unnatural amino acid (e.g., an unnatural amino acid contemplated herein) into a mammalian protein relative to a counterpart wild-type suppressor tRNA (in this context, a wild-type suppressor tRNA refers to a suppressor tRNA that corresponds to a wild-type tRNA molecule but for any modifications to the anti-codon region to impart suppression activity). The activity of the variant suppressor tRNA may be increased relative to the wild type suppressor tRNA, for example, by about 2.5 to about 200 fold, about 2.5 to about 150 fold, about 2.5 to about 100 fold about 2.5 to about 80 fold, about 2.5 to about 60 fold, about 2.5 to about 40 fold, about 2.5 to about 20 fold, about 2.5 to about 10 fold, about 2.5 to about 5 fold, about 5 to about 200 fold, about 5 to about 150 fold, about 5 to about 100 fold, about 5 to about 80 fold, about 5 to about 60 fold, about 5 to about 40 fold, about 5 to about 20 fold, about 5 to about 10 fold, about 10 to about 200 fold, about 10 to about 150 fold, about 10 to about 100 fold, about 10 to about 80 fold, about 10 to about 60 fold, about 10 to about 40 fold, about 10 to about 20 fold, about 20 to about 200 fold, about 20 to about 150 fold, about 20 to about 100 fold, about 20 to about 80 fold, about 20 to about 60 fold, about 20 to about 40 fold, about 40 to about 200 fold, about 40 to about 150 fold, about 40 to about 100 fold, about 40 to about 80 fold, about 40 to about 60 fold, about 60 to about 200 fold, about 60 to about 150 fold, about 60 to about 100 fold, about 60 to about 80 fold, about 80 to about 200 fold, about 80 to about 150 fold, about 80 to about 100 fold, about 100 to about 200 fold, about 100 to about 150 fold, or about 150 to about 200 fold.

It is contemplated that the tRNA may function in vitro or in vivo and can be provided to a translation system (e.g., an in vitro translation system or a cell) as a mature tRNA (e.g., an aminoacylated tRNA), or as a polynucleotide that encodes the tRNA.

A tRNA may be derived from a bacterial source, e.g., Escherichia coli, Thermus thermophilus, or Bacillus stearothermphilus. A tRNA may also be derived from an archaeal source, e.g, from the Methanosarcinacaea or Desulfitobacterium families, any of the M. barkeri (Mb), M. alvus (Ma), M. mazei (Mm) or D. hafnisense (Dh) families, Methanobacterium thermoautotrophicum, Haloferax volcanii, Halobacterium species NRC-1, or Archaeoglobus fulgidus. In other embodiments, eukaryotic sources can also be used, for example, plants, algae, protists, fungi, yeasts, or animals (e.g., mammals, insects, arthropods, etc.).

In certain embodiments, the tRNA is derived from an E. coli leucyl tRNA and, for example, is preferentially charged with a leucine analog over the naturally-occurring leucine amino acid by an aminoacyl-tRNA synthetase derived from an E. coli leucyl-tRNA synthetase, e.g., an aminoacyl-tRNA synthetase contemplated herein.

For example, the tRNA may comprise, consist essentially of, or consist of the nucleotide sequence of any one of SEQ ID NOs: 16-43, or a nucleotide sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOs: 16-43.

In certain embodiments, the tRNA is derived from an E. coli tryptophanyl tRNA and, for example, is preferentially charged with a tryptophan analog over the naturally-occurring tryptophan amino acid by an aminoacyl-tRNA synthetase derived from an E. coli tryptophanyl-tRNA synthetase, e.g., an aminoacyl-tRNA synthetase contemplated herein.

For example, the tRNA may comprise, consist essentially of, or consist of the nucleotide sequence of any one of SEQ ID NOs: 49-54 or 108-113, or a nucleotide sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOs: 49-54 or 108-113.

In certain embodiments, the tRNA is derived from an E. coli tyrosyl tRNA and, for example, is preferentially charged with a tyrosine analog over the naturally-occurring tyrosine amino acid by an aminoacyl-tRNA synthetase derived from an E. coli tyrosyl-tRNA synthetase, e.g., an aminoacyl-tRNA synthetase contemplated herein.

For example, the tRNA may comprise, consist essentially of, or consist of the nucleotide sequence of any one of SEQ ID NOs: 68-69 or 104-105, or a nucleotide sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOs: 68-69 or 104-105.

In certain embodiments, the tRNA is derived from a M. barkeri pyrrolysyl tRNA and, for example, is preferentially charged with a pyrrolysine analog over the naturally-occurring pyrrolysine amino acid by an aminoacyl-tRNA synthetase derived from a M. barkeri pyrrolysyl-tRNA synthetase, e.g., an aminoacyl-tRNA synthetase contemplated herein.

For example, the tRNA may comprise, consist essentially of, or consist of the nucleotide sequence of any one of SEQ ID NOs: 72-100 or 106-107, or a nucleotide sequence that has at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to any one of SEQ ID NOs: 72-100 or 106-107.

It is understood that, throughout the description, in each instance where a tRNA comprises, consists essentially of, or consists of a nucleotide sequence including one or more thymines (T), a tRNA is also contemplated that comprises, consists essentially of, or consists of the same nucleotide sequence including a uracil (U) in place of one or more of the thymines (T), or a uracil (U) in place of all the thymines (T). Similarly, in each instance where a tRNA comprises, consists essentially of, or consists of a nucleotide sequence including one or more uracils (U), a tRNA is also contemplated that comprises, consists essentially of, or consists of a nucleotide sequence including a thymine (T) in place of the one or more of the uracils (U), or a thymine (T) in place of all the uracils (U). In addition, additional modifications to the bases can be present.

Methods for producing recombinant tRNA are described in U.S. Patent Application Publication Nos. 2003/0108885 and 2005/0009049, Forster et al. (2003) PROC. NATL. ACAD. SCI. USA 100(11):6353-6357, and Feng et al. (2003), PROC. NATL. ACAD. SCI. USA 100(10): 5676-5681.

A tRNA may be aminoacylated (i.e., charged) with a desired unnatural amino acid (UAA) by any method, including enzymatic or chemical methods.

Enzymatic molecules capable of charging a tRNA include aminoacyl-tRNA synthetases, e.g., aminoacyl-tRNA synthetases disclosed herein. Additional enzymatic molecules capable of charging tRNA include ribozymes, for example, as described in Illangakekare et al. (1995) SCIENCE 267:643-647, Lohse et al. (1996) NATURE 381:442-444, Murakami et al. (2003) CHEMISTRY AND BIOLOGY 10:1077-1084, U.S. Patent Application Publication No. 2003/0228593,

Chemical aminoacylation methods include those described in Hecht (1992) ACC. CHEM. RES. 25:545, Heckler et al. (1988) BIOCHEM. 1988, 27:7254, Hecht et al. (1978) J. BIOL. CHEM. 253:4517, Cornish et al. (1995) ANGEW. CHEM. INT. ED. ENGL. 34:621, Robertson et al. (1991) J. AM. CHEM. SOC. 113:2722, Noren et al. (1989) SCIENCE 244:182, Bain et al. (1989) J. AM. CHEM. SOC. 111:8013, Bain et al. (1992) NATURE 356:537, Gallivan et al. (1997) CHEM. BIOL. 4:740, Turcatti et al. (1996) J. BIOL. CHEM. 271:19991, Nowak et al. (1995) SCIENCE 268:439, Saks et al. (1996) J. BIOL. CHEM. 271:23169, and Hohsaka et al. (1999) J. AM. CHEM. SOC. 121:34.

III. Unnatural Amino Acids (UAAs)

The invention relates to unnatural amino acids (UAAs) and their incorporation into proteins.

As used herein, an unnatural amino acid refers to any amino acid, modified amino acid, or amino acid analogue other than the following twenty genetically encoded alpha-amino acids: alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, valine. See, e.g., Biochemistry by L. Stryer, 3rd ed. 1988, Freeman and Company, New York, for structures of the twenty natural amino acids. The term unnatural amino acid also includes amino acids that occur by modification (e.g. post-translational modifications) of a natural amino acid but are not themselves naturally incorporated into a growing polypeptide chain by the translation complex.

Because unnatural amino acids typically differ from natural amino acids only in the structure of the side chain, unnatural amino acids may, for example, form amide bonds with other amino acids in the same manner in which they are formed in naturally occurring proteins. However, the unnatural amino acids have side chain groups that distinguish them from the natural amino acids. For example, the side chain may comprise an alkyl-, aryl-, acyl-, keto-, azido-, hydroxyl-, hydrazine, cyano-, halo-, hydrazide, alkenyl, alkyl, ether, thiol, seleno-, sulfonyl-, borate, boronate, phospho, phosphono, phosphine, heterocyclic, enone, imine, aldehyde, ester, thioacid, hydroxylamine, amine, and the like, or any combination thereof. Other non-naturally occurring amino acids include, but are not limited to, amino acids comprising a photoactivatable cross-linker, spin-labeled amino acids, fluorescent amino acids, metal binding amino acids, metal-containing amino acids, radioactive amino acids, amino acids with novel functional groups, amino acids that covalently or noncovalently interact with other molecules, photocaged and/or photoisomerizable amino acids, amino acids comprising biotin or a biotin analogue, glycosylated amino acids such as a sugar substituted serine, other carbohydrate modified amino acids, keto-containing amino acids, amino acids comprising polyethylene glycol or polyether, heavy atom substituted amino acids, chemically cleavable and/or photocleavable amino acids, amino acids with an elongated side chains as compared to natural amino acids, including but not limited to, polyethers or long chain hydrocarbons, including but not limited to, greater than about 5 or greater than about 10 carbons, carbon-linked sugar-containing amino acids, redox-active amino acids, amino thioacid containing amino acids, and amino acids comprising one or more toxic moiety.

In addition to unnatural amino acids that contain novel side chains, unnatural amino acids also optionally comprise modified backbone structures.

Many unnatural amino acids are based on natural amino acids, such as tyrosine, glutamine, phenylalanine, and the like. Tyrosine analogs include para-substituted tyrosines, ortho-substituted tyrosines, and meta substituted tyrosines, wherein the substituted tyrosine comprises a keto group (including but not limited to, an acetyl group), a benzoyl group, an amino group, a hydrazine, an hydroxyamine, a thiol group, a carboxy group, an isopropyl group, a methyl group, a C6-C20 straight chain or branched hydrocarbon, a saturated or unsaturated hydrocarbon, an O-methyl group, a polyether group, a nitro group, or the like. In addition, multiply substituted aryl rings are also contemplated. Glutamine analogs include, but are not limited to, α-hydroxy derivatives, γ-substituted derivatives, cyclic derivatives, and amide substituted glutamine derivatives. Exemplary phenylalanine analogs include, but are not limited to, para-substituted phenylalanines, ortho-substituted phenylalanines, and meta-substituted phenylalanines, wherein the substituent comprises a hydroxy group, a methoxy group, a methyl group, an allyl group, an aldehyde, an azido, an iodo, a bromo, a keto group (including but not limited to, an acetyl group), or the like. Specific examples of unnatural amino acids include, but are not limited to, a p-acetyl-L-phenylalanine, a p-propargyl-phenylalanine, O-methyl-L-tyrosine, an L-3-(2-naphthyl)alanine, a 3-methyl-phenylalanine, an O-4-allyl-L-tyrosine, a 4-propyl-L-tyrosine, a tri-O-acetyl-GcNAcβ-serine, an L-Dopa, a fluorinated phenylalanine, an isopropyl-L-phenylalanine, a p-azido-L-phenylalanine, a p-acyl-L-phenylalanine, a p-benzoyl-L-phenylalanine, an L-phosphoserine, a phosphonoserine, a phosphonotyrosine, a p-iodo-phenylalanine, a p-bromophenylalanine, a p-amino-L-phenylalanine, an isopropyl-L-phenylalanine, and a p-propargyloxy-phenylalanine, and the like.

Examples of structures of a variety of unnatural amino acids are provided in U.S. Patent Application Publication Nos. 2003/0082575 and 2003/0108885, PCT Publication No. WO 2002/085923, and Kiick et al. (2002) PROC. NATL. ACAD. SCI. USA 99:19-24.

In certain embodiments, an unnatural amino acid (UAA) comprises a bioconjugation handle. In certain embodiments, a method disclosed herein can be used to site-specifically incorporate two different UAAs, each with a different bioconjugation handle, into a single protein. In certain embodiments, the two bioconjugation handles can be chosen such that they each can be chemoselectively conjugated to two different labels using mutually orthogonal conjugation chemistries. Such pairs of bioconjugation handles include, for example: azide and alkyne, azide and ketone/aldehyde, azide and cyclopropene, ketone/aldehyde and cyclopropene, 5-hydroxyindole and azide, 5-hydroxyindole and cyclopropene, and 5-hydroxyindole and ketone/aldehyde,

An unnatural amino acid in a polypeptide may be used to attach another molecule to the polypeptide, including but not limited to, a label, a dye, a polymer, a water-soluble polymer, a stabilizing agent (e.g., a derivative of polyethylene glycol, a photoactivatable crosslinker, a radionuclide, a cytotoxic compound, a drug, an affinity label, a photoaffinity label, a reactive compound, a resin, a second protein or polypeptide or polypeptide analog, an antibody or antibody fragment, a metal chelator, a cofactor, a fatty acid, a carbohydrate, a polynucleotide, a DNA, a RNA, an antisense polynucleotide, a saccharide, a water-soluble dendrimer, a cyclodextrin, an inhibitory ribonucleic acid (e.g., a small interfering RNA (siRNA), a small nuclear RNA (snRNA), or a non-coding RNA), a biomaterial, a nanoparticle, a spin label, a fluorophore, a metal-containing moiety, a radioactive moiety, a novel functional group, a group that covalently or noncovalently interacts with other molecules, a photocaged moiety, an actinic radiation excitable moiety, a photoisomerizable moiety, biotin, a derivative of biotin, a biotin analogue, a moiety incorporating a heavy atom, a chemically cleavable group, a photocleavable group, an elongated side chain, a carbon-linked sugar, a redox-active agent, an amino thioacid, a toxic moiety, an isotopically labeled moiety, a biophysical probe or biochemical probe (e.g., a PET probe, a fluorescent probe or an EPR probe), a phosphorescent group, a chemiluminescent group, an electron dense group, a magnetic group, an intercalating group, a chromophore, an energy transfer agent, a biologically active agent, a detectable label (e.g., for analysis of uptake in viable cells versus non-viable cells), a small molecule, a quantum dot, a nanotransmitter, an immunomodulatory molecule, a targeting agent, a lipid based structure (e.g., a lipid-based nanoparticle), a microsphere, or any combination of the above.

Any suitable unnatural amino acid can be used with the methods described herein for incorporation into a protein of interest.

The unnatural amino acid may be a leucine analog (also referred to herein as a derivative). In certain embodiments, the leucine analog is a non-naturally occurring leucine analog. The invention provides a leucine analog depicted in FIG. 2A, or a composition comprising the leucine analog. For example, Formula A in FIG. 2A depicts an amino acid analog containing a side chain including a carbon containing chain n units (0-20 units) long. An O, S, CH2, or NH is present in at position X, and another carbon containing chain of n units (0-20 units) long can follow. A functional group Y is attached to the terminal carbon of second carbon containing chain (for example, functional groups 1-12 as depicted in FIG. 2A, where R represents a linkage to the terminal carbon atom the second carbon containing side chain). In one example, these functional groups can be used for bioconjugation of any amenable ligand to any protein of interest that is amenable to site-specific UAA incorporation. Formula B in FIG. 2A depicts a similar amino acid analog containing an side chains denoted as either Z—Y2 or Z—Y3 attached to the second carbon containing chain or the first carbon containing chain, respectively. Z represents a carbon chain comprising (CH2)n units, where n is any integer from 0-20. Y2 or Y3, independently, can be the same or different groups as those of Yi. The invention also provides a leucine analog depicted in FIG. 2B (LCA, LKET, or ACA), or a composition comprising the leucine analog depicted in FIG. 2B. Additional exemplary leucine analogs include those selected from linear alkyl halides and linear aliphatic chains comprising a functional group, for example, an alkyne, azide, cyclopropene, alkene, ketone, aldehyde, diazirine, or tetrazine functional group, as well as structures 1-6 shown in FIG. 2C. However, it is contemplated that the amino and carboxylate groups both attached to the first carbon of any amino acid shown in FIGS. 2A-2C would constitute portions of peptide bonds when the leucine analog is incorporated into a protein or polypeptide chain.

In addition, the leucine analogs set forth in FIG. 4A, referred to as C5AzMe and LCA can be used in the practice of the invention.

C5AzMe (Compound 5 as shown in FIG. 4B) can be prepared in a manner similar to the synthesis outlined in FIG. 4B. Compound 5 can be furnished by, for example, the deprotection of Compound 4. Deprotection of Compound 4 comprises the removal of a protecting group (e.g. Boc). Conditions for deprotection may include, but are not limited to, HCl in DCM. Compound 4 can be generated, for example, via nucleophilic substitution of Compound 3 when exposed to a suitable nucleophile (e.g. N3). Exemplary conditions for nucleophilic substitution include, but are not limited to, NaN3 in DMF at 80° C. Compound 3 can be prepared, for example, via nucleophilic addition of Compound 1 to Compound 2. Exemplary conditions for nucleophilic addition include, but are not limited to, K2CO3 at 0° C. to RT. Furthermore, if desired, the ester of Compound 5 can be removed by exposure to mild aqueous basic conditions to produce the carboxylic acid form of the UAA.

LCA (Compound 21 as shown in FIG. 4F) can be prepared in a manner similar to the synthesis outlined in FIG. 4F. Compound 21 can be prepared, for example, from Compound 20 through exposure of Compound 20 to a suitable acid, for example, but not limited to, 4M HCl in dioxane. Compound 20 can be generated through hydrolyzation of imine 19. Hydrolyzation of imine 19 can be accomplished, for example, using 1M HCl (aq.) in THF. Compound 19 can be generated, for example, via nucleophilic substitution of Compound 18 when exposed to a suitable nucleophile (e.g. N3). Exemplary conditions for nucleophilic substitution include, but are not limited to, NaN3 in DMF. Compound 18 can be prepared via nucleophilic addition of the enolate of Compound 16 to Compound 17. Suitable conditions for accomplishing synthesis of compound from Compounds 16 and 17 include, but are not limited to, tetrabutylammonium hydrogensulfate (TBAHS) and 10% NaOH in DCM. Additional methods for synthesis of LCA are shown in FIGS. 4D and 4E.

In certain embodiments, the unnatural amino acid is a tryptophan analog (also referred to herein as a derivative). In certain embodiments, the tryptophan analog is a non-naturally occurring tryptophan analog. Exemplary tryptophan analogs include 5-azidotryptophan, 5-propargyloxytryptophan, 5-aminotryptophan, 5-methoxytryptophan, 5-O-allyltryptophan or 5-bromotryptophan. Additional exemplary tryptophan analogs are depicted in FIG. 3. However, it is contemplated that the amino and carboxylate groups both attached to the first carbon of the tryptophan analogs in FIG. 3 would constitute portions of peptide bonds when the tryptophan analog is incorporated into a protein or polypeptide chain.

In addition, the tryptophan analog set forth in FIG. 4A, referred to as AzW, can be used in the practice of the invention.

AzW (Compound 15 as shown in FIG. 4C) can be prepared in a manner similar to the synthesis outlined in FIG. 4C. Compound 15 can be prepared, for example, under basic conditions from its hydrochloride salt 14. Exemplary basic conditions include, but are not limited to, KOtBu in THF. Hydrochloride salt 14 can be prepared, for example, via saponification followed by deprotection of Compound 13. Conditions for saponification and deprotection of a protecting group (e.g., Boc) are known to a person of ordinary skill in the art. For example, saponification can be accomplished using 1M NaOH in MeOH. In certain embodiments, conditions for deprotection include, but are not limited to, HCl (aq.). Compound 13 can be synthesized, for example, through a metal-mediated coupling of Compound 12 with a suitable azide source. Compound 13 can be made, for example, from Compound 12 using NaN3, Cu(OAc)2 in MeOH. Compound 12 can, for example, be prepared from Compound 11 through metal-catalyzed boronation of Compound 11. Exemplary conditions for metal-catalyzed boronation include, but are not limited to B2pin2, PdCl2.dppf, and KOAc in 1,4-dioxane. Compound 11 can be prepared, for example, via protection of Compound 10 using a suitable protecting group (e.g., Boc). Protection of Compound 10 can be accomplished using Boc2O, Et3N, and DMAP in CH2Cl2. Compound 10 can be synthesized, for example, from Compound 9 under conditions suitable for reducing an oxime, for example, using Zn in AcOH. Compound 9 can synthesized, for example, via nucleophilic addition of indole 8 to Compound 7. Narcoleptic addition of Compound 8 to Compound 7 can occur in the presence of Na2CO3 in CH2Cl2. Compound 7 can be prepared, for example, by exposing Compound 6 to hydroxylamine hydrochloride in methanol.

In certain embodiments, the unnatural amino acid is a tyrosine analog (also referred to herein as a derivative). In certain embodiments, the tyrosine analog is a non-naturally occurring tyrosine analog. Exemplary tyrosine analogs include o-methyltyrosine (OmeY), p-azidophenylalanine (AzF), o-propargyltyrosine (OpropY or PrY), and p-acetylphenylalanine (AcF). Exemplary tryptophan analogs are depicted in FIG. 5.

In certain embodiments, the unnatural amino acid is a pyrrolysine analog (also referred to herein as a derivative). In certain embodiments, the pyrrolysine analog is a non-naturally occurring pyrrolysine analog. Exemplary pyrrolysine analogs include aminocaprylic acid (Cap), H-Lys(Boc)-OH (Boc-Lysine, BocK), azidolysine (AzK), H-propargyl-lysine (hPrK), and cyclopropenelysine (CpK). Exemplary pyrrolysine analogs are depicted in FIG. 6.

Many unnatural amino acids are commercially available, e.g., from Sigma-Aldrich (St. Louis, Mo., USA), Novabiochem (Darmstadt, Germany), or Peptech (Burlington, Mass., USA). Those that are not commercially available can be synthesized using standard methods known to those of ordinary skill in the art. For organic synthesis techniques, see, e.g., Organic Chemistry by Fessendon and Fessendon, (1982, Second Edition, Willard Grant Press, Boston Mass.); Advanced Organic Chemistry by March (Third Edition, 1985, Wiley and Sons, New York); and Advanced Organic Chemistry by Carey and Sundberg (Third Edition, Parts A and B, 1990, Plenum Press, New York). Additional exemplary publications describing the synthesis of unnatural amino acids appear in PCT Publication No. WO2002/085923, U.S. Patent Application Publication No. 2004/0198637, Matsoukas et al. (1995) J. MED. CHEM. 38:4660-4669, King et al. (1949) J. CHEM. SOC. 3315-3319, Friedman et al. (1959) J. AM. CHEM. SOC. 81:3750-3752, Craig et al. (1988) J. ORG. CHEM. 53:1167-1170, Azoulay et al. (1991) EUR. J. MED. CHEM. 26:201-5, Koskinen et al. (1989) J. ORG. CHEM. 54:1859-1866, Christie et al. (1985) J. ORG. CHEM. 50:1239-1246, Barton et al. (1987) TETRAHEDRON 43:4297-4308, and Subasinghe et al. (1992) J. MED. CHEM. 35:4602-7.

IV. Vectors

tRNAs, aminoacyl-tRNA synthetases, or any other molecules of interest may be expressed in a cell of interest by incorporating a gene encoding the molecule into an appropriate expression vector. As used herein, “expression vector” refers to a vector comprising a recombinant polynucleotide comprising expression control sequences operatively linked to a nucleotide sequence to be expressed. An expression vector comprises sufficient cis-acting elements for expression; other elements for expression can be supplied by the host cell or in an in vitro expression system.

tRNAs, aminoacyl-tRNA synthetases, or any other molecules of interest may be introduced to a cell of interest by incorporating a gene encoding the molecule into an appropriate transfer vector. The term “transfer vector” refers to a vector comprising a recombinant polynucleotide which can be used to deliver the polynucleotide to the interior of a cell. It is understood that a vector may be both an expression vector and a transfer vector.

Vectors (e.g., expression vectors or transfer vectors) include all those known in the art, such as cosmids, plasmids (e.g., naked or contained in liposomes), retrotransposons (e.g. piggyback, sleeping beauty), and viruses (e.g., lentiviruses, retroviruses, adenoviruses, and adeno-associated viruses) that incorporate the recombinant polynucleotide of interest.

Typical vectors contain transcription and translation terminators, transcription and translation initiation sequences, and promoters useful for regulation of the expression of the particular target nucleic acid. The vectors optionally comprise generic expression cassettes containing at least one independent terminator sequence, sequences permitting replication of the cassette in eukaryotes, or prokaryotes, or both (including but not limited to, shuttle vectors) and selection markers for both prokaryotic and eukaryotic systems.

In certain embodiments, the vector comprises a regulatory sequence or promoter operably linked to the nucleotide sequence encoding the suppressor tRNA and/or the tRNA synthetase. The term “operably linked” refers to a linkage of polynucleotide elements in a functional relationship. A nucleic acid sequence is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. For instance, a promoter or enhancer is operably linked to a gene if it affects the transcription of the gene. Operably linked nucleotide sequences are typically contiguous. However, as enhancers generally function when separated from the promoter by several kilobases and intronic sequences may be of variable lengths, some polynucleotide elements may be operably linked but not directly flanked and may even function in trans from a different allele or chromosome.

Exemplary promoters which may be employed include, but are not limited to, the retroviral LTR, the SV40 promoter, the human cytomegalovirus (CMV) promoter, the U6 promoter, the EF1α promoter, the CAG promoter, the H1 promoter, the UbiC promoter, the PGK promoter, the 7SK promoter, a pol II promoter, a pol III promoter, or any other promoter (e.g., cellular promoters such as eukaryotic cellular promoters including, but not limited to, the histone, pol III, and β-actin promoters). Other viral promoters which may be employed include, but are not limited to, adenovirus promoters, TK promoters, and B19 parvovirus promoters. The selection of a suitable promoter will be apparent to those skilled in the art from the teachings contained herein. In certain embodiments, a vector comprises a nucleotide sequence encoding an aminoacyl-tRNA synthetase operably linked to a CMV or an EF1α promoter and/or a nucleotide sequence encoding a suppressor tRNA operably linked to a U6 or an H1 promoter.

In certain embodiments, the vector is a viral vector. The term “virus” is used herein to refer to an obligate intracellular parasite having no protein-synthesizing or energy-generating mechanism. Exemplary viral vectors include retroviral vectors (e.g., lentiviral vectors), adenoviral vectors, adeno-associated viral vectors, herpesviruses vectors, epstein-barr virus (EBV) vectors, polyomavirus vectors (e.g., simian vacuolating virus 40 (SV40) vectors), poxvirus vectors, and pseudotype virus vectors.

The virus may be a RNA virus (having a genome that is composed of RNA) or a DNA virus (having a genome composed of DNA). In certain embodiments, the viral vector is a DNA virus vector. Exemplary DNA viruses include parvoviruses (e.g., adeno-associated viruses). adenoviruses, asfarviruses, herpesviruses (e.g., herpes simplex virus 1 and 2 (HSV-1 and HSV-2), epstein-barr virus (EBV), cytomegalovirus (CMV)), papillomoviruses (e.g., HPV), polyomaviruses (e.g., simian vacuolating virus 40 (SV40)), and poxviruses (e.g., vaccinia virus, cowpox virus, smallpox virus, fowlpox virus, sheeppox virus, myxoma virus). In certain embodiments, the viral vector is a RNA virus vector. Exemplary RNA viruses include bunyaviruses (e.g., hantavirus), coronaviruses, flaviviruses (e.g., yellow fever virus, west nile virus, dengue virus), hepatitis viruses (e.g., hepatitis A virus, hepatitis C virus, hepatitis E virus), influenza viruses (e.g., influenza virus type A, influenza virus type B, influenza virus type C), measles virus, mumps virus, noroviruses (e.g., Norwalk virus), poliovirus, respiratory syncytial virus (RSV), retroviruses (e.g., human immunodeficiency virus-1 (HIV-1)) and toroviruses.

Adeno-Associated Virus (AAV) Vectors

In certain embodiments, a vector is an adeno-associated virus (AAV) vector. AAV is a small, nonenveloped icosahedral virus of the genus Dependoparvovirus and family Parvovirus. AAV has a single-stranded linear DNA genome of approximately 4.7 kb. AAV is capable of infecting both dividing and quiescent cells of several tissue types, with different AAV serotypes exhibiting different tissue tropism.

AAV includes numerous serologically distinguishable types including serotypes AAV-1 to AAV-12, as well as more than 100 serotypes from nonhuman primates (See, e.g., Srivastava (2008) J. CELL BIOCHEM., 105(1): 17-24, and Gao et al. (2004) J. VIROL., 78(12), 6381-6388). The serotype of the AAV vector used in the present invention can be selected by a skilled person in the art based on the efficiency of delivery, tissue tropism, and immunogenicity. For example, AAV-1, AAV-2, AAV-4, AAV-5, AAV-8, and AAV-9 can be used for delivery to the central nervous system; AAV-1, AAV-8, and AAV-9 can be used for delivery to the heart; AAV-2 can be used for delivery to the kidney; AAV-7, AAV-8, and AAV-9 can be used for delivery to the liver; AAV-4, AAV-5, AAV-6, AAV-9 can be used for delivery to the lung, AAV-8 can be used for delivery to the pancreas, AAV-2, AAV-5, and AAV-8 can be used for delivery to the photoreceptor cells; AAV-1, AAV-2, AAV-4, AAV-5, and AAV-8 can be used for delivery to the retinal pigment epithelium; AAV-1, AAV-6, AAV-7, AAV-8, and AAV-9 can be used for delivery to the skeletal muscle. In certain embodiments, the AAV capsid protein comprises a sequence as disclosed in U.S. Pat. No. 7,198,951, such as, but not limited to, AAV-9 (SEQ ID NOs: 1-3 of U.S. Pat. No. 7,198,951), AAV-2 (SEQ ID NO: 4 of U.S. Pat. No. 7,198,951), AAV-1 (SEQ ID NO: 5 of U.S. Pat. No. 7,198,951), AAV-3 (SEQ ID NO: 6 of U.S. Pat. No. 7,198,951), and AAV-8 (SEQ ID NO: 7 of U.S. Pat. No. 7,198,951). AAV serotypes identified from rhesus monkeys, e.g., rh.8, rh.10, rh.39, rh.43, and rh.74, are also contemplated in the instant invention. Besides the natural AAV serotypes, modified AAV capsids have been developed for improving efficiency of delivery, tissue tropism, and immunogenicity. Exemplary natural and modified AAV capsids are disclosed in U.S. Pat. Nos. 7,906,111, 9,493,788, and 7,198,951, and PCT Publication No. WO2017189964A2.

The wild-type AAV genome contains two 145 nucleotide inverted terminal repeats (ITRs), which contain signal sequences directing AAV replication, genome encapsidation and integration. In addition to the ITRs, three AAV promoters, p5, p19, and p40, drive expression of two open reading frames encoding rep and cap genes. Two rep promoters, coupled with differential splicing of the single AAV intron, result in the production of four rep proteins (Rep 78, Rep 68, Rep 52, and Rep 40) from the rep gene. Rep proteins are responsible for genomic replication. The Cap gene is expressed from the p40 promoter, and encodes three capsid proteins (VP1, VP2, and VP3) which are splice variants of the cap gene. These proteins form the capsid of the AAV particle.

Because the cis-acting signals for replication, encapsidation, and integration are contained within the ITRs, some or all of the 4.3 kb internal genome may be replaced with foreign DNA, for example, an expression cassette for an exogenous gene of interest. Accordingly, in certain embodiments, the AAV vector comprises a genome comprising an expression cassette for an exogenous gene flanked by a 5′ ITR and a 3′ ITR. The ITRs may be derived from the same serotype as the capsid or a derivative thereof. Alternatively, the ITRs may be of a different serotype from the capsid, thereby generating a pseudotyped AAV. In certain embodiments, the ITRs are derived from AAV-2. In certain embodiments, the ITRs are derived from AAV-5. At least one of the ITRs may be modified to mutate or delete the terminal resolution site, thereby allowing production of a self-complementary AAV vector.

The rep and cap proteins can be provided in trans, for example, on a plasmid, to produce an AAV vector. A host cell line permissive of AAV replication must express the rep and cap genes, the ITR-flanked expression cassette, and helper functions provided by a helper virus, for example adenoviral genes Ela, E1b55K, E2a, E4orf6, and VA (Weitzman et al., Adeno-associated virus biology. Adeno-Associated Virus: Methods and Protocols, pp. 1-23, 2011). Methods for generating and purifying AAV vectors have been described in detail (See e.g., Mueller et al., (2012) CURRENT PROTCOLS IN MICROBIOLOGY, 14D.1.1-14D.1.21, Production and Discovery of Novel Recombinant Adeno-Associated Viral Vectors). Numerous cell types are suitable for producing AAV vectors, including HEK293 cells, COS cells, HeLa cells, BHK cells, Vero cells, as well as insect cells (See e.g. U.S. Pat. Nos. 6,156,303, 5,387,484, 5,741,683, 5,691,176, 5,688,676, and 8,163,543, U.S. Patent Publication No. 20020081721, and PCT Publication Nos. WO00/47757, WO00/24916, and WO96/17947). AAV vectors are typically produced in these cell types by one plasmid containing the ITR-flanked expression cassette, and one or more additional plasmids providing the additional AAV and helper virus genes.

AAV of any serotype may be used in the present invention. Similarly, it is contemplated that any adenoviral type may be used, and a person of skill in the art will be able to identify AAV and adenoviral types suitable for the production of their desired recombinant AAV vector (rAAV). AAV particles may be purified, for example by affinity chromatography, iodixonal gradient, or CsCl gradient.

AAV vectors may have single-stranded genomes that are 4.7 kb in size, or are larger or smaller than 4.7 kb, including oversized genomes that are as large as 5.2 kb, or as small as 3.0 kb. Thus, where the exogenous gene of interest to be expressed from the AAV vector is small, the AAV genome may comprise a stuffer sequence. Further, vector genomes may be substantially self-complementary thereby allowing for rapid expression in the cell. In certain embodiments, the genome of a self-complementary AAV vector comprises from 5′ to 3′: a 5′ ITR; a first nucleic acid sequence comprising a promoter and/or enhancer operably linked to a coding sequence of a gene of interest; a modified ITR that does not have a functional terminal resolution site; a second nucleic acid sequence complementary or substantially complementary to the first nucleic acid sequence; and a 3′ ITR. AAV vectors containing genomes of all types are suitable for use in the method of the present invention.

Non-limiting examples of AAV vectors include pAAV-MCS (Agilent Technologies), pAAVK-EF1α-MCS (System Bio Catalog # AAV502A-1), pAAVK-EF1α-MCS1-CMV-MCS2 (System Bio Catalog # AAV503A-1), pAAV-ZsGreenl (Clontech Catalog #6231), pAAV-MCS2 (Addgene Plasmid #46954), AAV-Stuffer (Addgene Plasmid #106248), pAAVscCBPIGpluc (Addgene Plasmid #35645), AAVS1_Puro_PGK1_3×FLAG Twin_Strep (Addgene Plasmid #68375), pAAV-RAM-d2TTA::TRE-MCS-WPRE-pA (Addgene Plasmid #63931), pAAV-UbC (Addgene Plasmid #62806), pAAVS1-P-MCS (Addgene Plasmid #80488), pAAV-Gateway (Addgene Plasmid #32671), pAAV-Puro_siKD (Addgene Plasmid #86695), pAAVS1-Nst-MCS (Addgene Plasmid #80487), pAAVS1-Nst-CAG-DEST (Addgene Plasmid #80489), pAAVS1-P-CAG-DEST (Addgene Plasmid #80490), pAAVf-EnhCB-lacZnls (Addgene Plasmid #35642), and pAAVS1-shRNA (Addgene Plasmid #82697). These vectors can be modified to be suitable for therapeutic use. For example, an exogenous gene of interest can be inserted in a multiple cloning site, and a selection marker (e.g., puro or a gene encoding a fluorescent protein) can be deleted or replaced with another (same or different) exogenous gene of interest. Further examples of AAV vectors are disclosed in U.S. Pat. Nos. 5,871,982, 6,270,996, 7,238,526, 6,943,019, 6,953,690, 9,150,882, and 8,298,818, U.S. Patent Publication No. 2009/0087413, and PCT Publication Nos. WO2017075335A1, WO2017075338A2, and WO2017201258A1.

Lentivirus Vectors

In certain embodiments, the viral vector can be a retroviral vector. Examples of retroviral vectors include moloney murine leukemia virus vectors, spleen necrosis virus vectors, and vectors derived from retroviruses such as rous sarcoma virus, harvey sarcoma virus, avian leukosis virus, human immunodeficiency virus, myeloproliferative sarcoma virus, and mammary tumor virus. Retroviral vectors are useful as agents to mediate retroviral-mediated gene transfer into eukaryotic cells.

In certain embodiments, the retroviral vector is a lentiviral vector. Exemplary lentiviral vectors include vectors derived from human immunodeficiency virus-1 (HIV-1), human immunodeficiency virus-2 (HIV-2), simian immunodeficiency virus (SIV), feline immunodeficiency virus (FIV), bovine immunodeficiency virus (BIV), Jembrana Disease Virus (JDV), equine infectious anemia virus (EIAV), and caprine arthritis encephalitis virus (CAEV).

Retroviral vectors typically are constructed such that the majority of sequences coding for the structural genes of the virus are deleted and replaced by the gene(s) of interest. Often, the structural genes (i.e., gag, pol, and env), are removed from the retroviral backbone using genetic engineering techniques known in the art. Accordingly, a minimum retroviral vector comprises from 5′ to 3′: a 5′ long terminal repeat (LTR), a packaging signal, an optional exogenous promoter and/or enhancer, an exogenous gene of interest, and a 3′ LTR. If no exogenous promoter is provided, gene expression is driven by the 5′ LTR, which is a weak promoter and requires the presence of Tat to activate expression. The structural genes can be provided in separate vectors for manufacture of the lentivirus, rendering the produced virions replication-defective. Specifically, with respect to lentivirus, the packaging system may comprise a single packaging vector encoding the Gag, Pol, Rev, and Tat genes, and a third, separate vector encoding the envelope protein Env (usually VSV-G due to its wide infectivity). To improve the safety of the packaging system, the packaging vector can be split, expressing Rev from one vector, Gag and Pol from another vector. Tat can also be eliminated from the packaging system by using a retroviral vector comprising a chimeric 5′ LTR, wherein the U3 region of the 5′ LTR is replaced with a heterologous regulatory element.

The genes can be incorporated into the proviral backbone in several general ways. The most straightforward constructions are ones in which the structural genes of the retrovirus are replaced by a single gene that is transcribed under the control of the viral regulatory sequences within the LTR. Retroviral vectors have also been constructed which can introduce more than one gene into target cells. Usually, in such vectors one gene is under the regulatory control of the viral LTR, while the second gene is expressed either off a spliced message or is under the regulation of its own, internal promoter.

Accordingly, the new gene(s) are flanked by 5′ and 3′ LTRs, which serve to promote transcription and polyadenylation of the virion RNAs, respectively. The term “long terminal repeat” or “LTR” refers to domains of base pairs located at the ends of retroviral DNAs which, in their natural sequence context, are direct repeats and contain U3, R and U5 regions. LTRs generally provide functions fundamental to the expression of retroviral genes (e.g., promotion, initiation and polyadenylation of gene transcripts) and to viral replication. The LTR contains numerous regulatory signals including transcriptional control elements, polyadenylation signals, and sequences needed for replication and integration of the viral genome. The U3 region contains the enhancer and promoter elements. The U5 region is the sequence between the primer binding site and the R region and contains the polyadenylation sequence. The R (repeat) region is flanked by the U3 and U5 regions. In certain embodiments, the R region comprises a trans-activation response (TAR) genetic element, which interacts with the trans-activator (tat) genetic element to enhance viral replication. This element is not required in embodiments wherein the U3 region of the 5′ LTR is replaced by a heterologous promoter.

In certain embodiments, the retroviral vector comprises a modified 5′ LTR and/or 3′ LTR. Modifications of the 3′ LTR are often made to improve the safety of lentiviral or retroviral systems by rendering viruses replication-defective. In specific embodiments, the retroviral vector is a self-inactivating (SIN) vector. As used herein, a SIN retroviral vector refers to a replication-defective retroviral vector in which the 3′ LTR U3 region has been modified (e.g., by deletion or substitution) to prevent viral transcription beyond the first round of viral replication. This is because the 3′ LTR U3 region is used as a template for the 5′ LTR U3 region during viral replication and, thus, the viral transcript cannot be made without the U3 enhancer-promoter. In a further embodiment, the 3′ LTR is modified such that the U5 region is replaced, for example, with an ideal polyadenylation sequence. It should be noted that modifications to the LTRs such as modifications to the 3′ LTR, the 5′ LTR, or both 3′ and 5′ LTRs, are also included in the invention.

In certain embodiments, the U3 region of the 5′ LTR is replaced with a heterologous promoter to drive transcription of the viral genome during production of viral particles. Examples of heterologous promoters which can be used include, for example, viral simian virus 40 (SV40) (e.g., early or late), cytomegalovirus (CMV) (e.g., immediate early), Moloney murine leukemia virus (MoMLV), Rous sarcoma virus (RSV), and herpes simplex virus (HSV) (thymidine kinase) promoters. Typical promoters are able to drive high levels of transcription in a Tat-independent manner. This replacement reduces the possibility of recombination to generate replication-competent virus, because there is no complete U3 sequence in the virus production system.

Adjacent the 5′ LTR are sequences necessary for reverse transcription of the genome (the tRNA primer binding site) and for efficient packaging of viral RNA into particles (the Psi site). As used herein, the term “packaging signal” or “packaging sequence” refers to sequences located within the retroviral genome which are required for encapsidation of retroviral RNA strands during viral particle formation (see e.g., Clever et al., 1995 J. VIROLOGY, 69(4):2101-09). The packaging signal may be a minimal packaging signal (also referred to as the psi [Ψ] sequence) needed for encapsidation of the viral genome.

In certain embodiments, the retroviral vector (e.g., lentiviral vector) further comprises a FLAP. As used herein, the term “FLAP” refers to a nucleic acid whose sequence includes the central polypurine tract and central termination sequences (cPPT and CTS) of a retrovirus, e.g., HIV-1 or HIV-2. Suitable FLAP elements are described in U.S. Pat. No. 6,682,907 and in Zennou et al. (2000) CELL, 101:173. During reverse transcription, central initiation of the plus-strand DNA at the cPPT and central termination at the CTS lead to the formation of a three-stranded DNA structure: a central DNA flap. While not wishing to be bound by any theory, the DNA flap may act as a cis-active determinant of lentiviral genome nuclear import and/or may increase the titer of the virus. In particular embodiments, the retroviral vector backbones comprise one or more FLAP elements upstream or downstream of the heterologous genes of interest in the vectors. For example, in particular embodiments, a transfer plasmid includes a FLAP element. In one embodiment, a vector of the invention comprises a FLAP element isolated from HIV-1.

In certain embodiments, the retroviral vector (e.g., lentiviral vector) further comprises an export element. In one embodiment, retroviral vectors comprise one or more export elements. The term “export element” refers to a cis-acting post-transcriptional regulatory element which regulates the transport of an RNA transcript from the nucleus to the cytoplasm of a cell. Examples of RNA export elements include, but are not limited to, the human immunodeficiency virus (HIV) RRE (see e.g., Cullen et al., (1991) J. VIROL. 65: 1053; and Cullen et al., (1991) CELL 58: 423) and the hepatitis B virus post-transcriptional regulatory element (HPRE). Generally, the RNA export element is placed within the 3′ UTR of a gene, and can be inserted as one or multiple copies.

In certain embodiments, the retroviral vector (e.g., lentiviral vector) further comprises a posttranscriptional regulatory element. A variety of posttranscriptional regulatory elements can increase expression of a heterologous nucleic acid, e.g., woodchuck hepatitis virus posttranscriptional regulatory element (WPRE; see Zufferey et al., (1999) J. VIROL., 73:2886); the posttranscriptional regulatory element present in hepatitis B virus (HPRE) (Huang et al., MOL. CELL. BIOL., 5:3864); and the like (Liu et al., (1995), GENES DEV., 9:1766). The posttranscriptional regulatory element is generally positioned at the 3′ end the heterologous nucleic acid sequence. This configuration results in synthesis of an mRNA transcript whose 5′ portion comprises the heterologous nucleic acid coding sequences and whose 3′ portion comprises the posttranscriptional regulatory element sequence. In certain embodiments, vectors of the invention lack or do not comprise a posttranscriptional regulatory element such as a WPRE or HPRE, because in some instances these elements increase the risk of cellular transformation and/or do not substantially or significantly increase the amount of mRNA transcript or increase mRNA stability. Therefore, in certain embodiments, vectors of the invention lack or do not comprise a WPRE or HPRE as an added safety measure.

Elements directing the efficient termination and polyadenylation of the heterologous nucleic acid transcripts increase heterologous gene expression. Transcription termination signals are generally found downstream of the polyadenylation signal. Accordingly, in certain embodiments, the retroviral vector (e.g., lentiviral vector) further comprises a polyadenylation signal. The term “polyadenylation signal” or “polyadenylation sequence” as used herein denotes a DNA sequence which directs both the termination and polyadenylation of the nascent RNA transcript by RNA polymerase H. Efficient polyadenylation of the recombinant transcript is desirable as transcripts lacking a polyadenylation signal are unstable and are rapidly degraded. Illustrative examples of polyadenylation signals that can be used in a vector of the invention, includes an ideal polyadenylation sequence (e.g., AATAAA, ATTAAA AGTAAA), a bovine growth hormone polyadenylation sequence (BGHpA), a rabbit β-globin polyadenylation sequence (rogpA), or another suitable heterologous or endogenous polyadenylation sequence known in the art.

In certain embodiments, a retroviral vector further comprises an insulator element. Insulator elements may contribute to protecting retrovirus-expressed sequences, e.g., therapeutic genes, from integration site effects, which may be mediated by cis-acting elements present in genomic DNA and lead to deregulated expression of transferred sequences (i.e., position effect; see, e.g., Burgess-Beusse et al., (2002) PROC. NATL. ACAD. SCI., USA, 99:16433; and Zhan et al., 2001, HUM. GENET., 109:471). In certain embodiments, the retroviral vector comprises an insulator element in one or both LTRs or elsewhere in the region of the vector that integrates into the cellular genome. Suitable insulators for use in the invention include, but are not limited to, the chicken β-globin insulator (see Chung et al., (1993). CELL 74:505; Chung et al., (1997) PROC. NATL. ACAD. SCI., USA 94:575; and Bell et al., 1999. CELL 98:387). Examples of insulator elements include, but are not limited to, an insulator from a β-globin locus, such as chicken HS4.

Non-limiting examples of lentiviral vectors include pLVX-EF1alpha-AcGFP1-C1 (Clontech Catalog #631984), pLVX-EF1alpha-IRES-mCherry (Clontech Catalog #631987), pLVX-Puro (Clontech Catalog #632159), pLVX-IRES-Puro (Clontech Catalog #632186), pLenti6N5-DEST™ (Thermo Fisher), pLenti6.2N5-DEST™ (Thermo Fisher), pLKO.1 (Plasmid #10878 at Addgene), pLKO.3G (Plasmid #14748 at Addgene), pSico (Plasmid #11578 at Addgene), pLJM1-EGFP (Plasmid #19319 at Addgene), FUGW (Plasmid #14883 at Addgene), pLVTHM (Plasmid #12247 at Addgene), pLVUT-tTR-KRAB (Plasmid #11651 at Addgene), pLL3.7 (Plasmid #11795 at Addgene), pLB (Plasmid #11619 at Addgene), pWPXL (Plasmid #12257 at Addgene), pWPI (Plasmid #12254 at Addgene), EF.CMV.RFP (Plasmid #17619 at Addgene), pLenti CMV Puro DEST (Plasmid #17452 at Addgene), pLenti-puro (Plasmid #39481 at Addgene), pULTRA (Plasmid #24129 at Addgene), pLX301 (Plasmid #25895 at Addgene), pHIV-EGFP (Plasmid #21373 at Addgene), pLV-mCherry (Plasmid #36084 at Addgene), pLionII (Plasmid #1730 at Addgene), pInducer10-mir-RUP-PheS (Plasmid #44011 at Addgene). These vectors can be modified to be suitable for therapeutic use. For example, a selection marker (e.g., puro, EGFP, or mCherry) can be deleted or replaced with a second exogenous gene of interest. Further examples of lentiviral vectors are disclosed in U.S. Pat. Nos. 7,629,153, 7,198,950, 8,329,462, 6,863,884, 6,682,907, 7,745,179, 7,250,299, 5,994,136, 6,287,814, 6,013,516, 6,797,512, 6,544,771, 5,834,256, 6,958,226, 6,207,455, 6,531,123, and 6,352,694, and PCT Publication No. WO2017/091786.

Adenoviral Vectors

In certain embodiments, the viral vector can be an adenoviral vector. Adenoviruses are medium-sized (90-100 nm), non-enveloped (naked), icosahedral viruses composed of a nucleocapsid and a double-stranded linear DNA genome. The term “adenovirus” refers to any virus in the genus Adenoviridiae including, but not limited to, human, bovine, ovine, equine, canine, porcine, murine, and simian adenovirus subgenera. Typically, an adenoviral vector is generated by introducing one or more mutations (e.g., a deletion, insertion, or substitution) into the adenoviral genome of the adenovirus so as to accommodate the insertion of a non-native nucleic acid sequence, for example, for gene transfer, into the adenovirus.

A human adenovirus can be used as the source of the adenoviral genome for the adenoviral vector. For instance, an adenovirus can be of subgroup A (e.g., serotypes 12, 18, and 31), subgroup B (e.g., serotypes 3, 7, 11, 14, 16, 21, 34, 35, and 50), subgroup C (e.g., serotypes 1, 2, 5, and 6), subgroup D (e.g., serotypes 8, 9, 10, 13, 15, 17, 19, 20, 22-30, 32, 33, 36-39, and 42-48), subgroup E (e.g., serotype 4), subgroup F (e.g., serotypes 40 and 41), an unclassified serogroup (e.g., serotypes 49 and 51), or any other adenoviral serogroup or serotype. Adenoviral serotypes 1 through 51 are available from the American Type Culture Collection (ATCC, Manassas, Va.). Non-group C adenoviral vectors, methods of producing non-group C adenoviral vectors, and methods of using non-group C adenoviral vectors are disclosed in, for example, U.S. Pat. Nos. 5,801,030, 5,837,511, and 5,849,561, and PCT Publication Nos. WO1997/012986 and WO1998/053087.

Non-human adenovirus (e.g., ape, simian, avian, canine, ovine, or bovine adenoviruses) can be used to generate the adenoviral vector (i.e., as a source of the adenoviral genome for the adenoviral vector). For example, the adenoviral vector can be based on a simian adenovirus, including both new world and old world monkeys (see, e.g., Virus Taxonomy: VHIth Report of the International Committee on Taxonomy of Viruses (2005)). A phylogeny analysis of adenoviruses that infect primates is disclosed in, e.g., Roy et al. (2009) PLOS PATHOG. 5(7):e1000503. A gorilla adenovirus can be used as the source of the adenoviral genome for the adenoviral vector. Gorilla adenoviruses and adenoviral vectors are described in, e.g., PCT Publication Nos.WO2013/052799, WO2013/052811, and WO2013/052832. The adenoviral vector can also comprise a combination of subtypes and thereby be a “chimeric” adenoviral vector.

The adenoviral vector can be replication-competent, conditionally replication-competent, or replication-deficient. A replication-competent adenoviral vector can replicate in typical host cells, i.e., cells typically capable of being infected by an adenovirus. A conditionally-replicating adenoviral vector is an adenoviral vector that has been engineered to replicate under pre-determined conditions. For example, replication-essential gene functions, e.g., gene functions encoded by the adenoviral early regions, can be operably linked to an inducible, repressible, or tissue-specific transcription control sequence, e.g., a promoter. Conditionally-replicating adenoviral vectors are further described in U.S. Pat. No. 5,998,205. A replication-deficient adenoviral vector is an adenoviral vector that requires complementation of one or more gene functions or regions of the adenoviral genome that are required for replication, as a result of, for example, a deficiency in one or more replication-essential gene function or regions, such that the adenoviral vector does not replicate in typical host cells, especially those in a human to be infected by the adenoviral vector.

Preferably, the adenoviral vector is replication-deficient, such that the replication-deficient adenoviral vector requires complementation of at least one replication-essential gene function of one or more regions of the adenoviral genome for propagation (e.g., to form adenoviral vector particles). The adenoviral vector can be deficient in one or more replication-essential gene functions of only the early regions (i.e., E1-E4 regions) of the adenoviral genome, only the late regions (i.e., L1-L5 regions) of the adenoviral genome, both the early and late regions of the adenoviral genome, or all adenoviral genes (i.e., a high capacity adenovector (HC-Ad)). See, e.g., Morsy et al. (1998) PROC. NATL. ACAD. SCI. USA 95: 965-976, Chen et al. (1997) PROC. NATL. ACAD. SCI. USA 94: 1645-1650, and Kochanek et al. (1999) HUM. GENE THER. 10(15):2451-9. Examples of replication-deficient adenoviral vectors are disclosed in U.S. Pat. Nos. 5,837,511, 5,851,806, 5,994,106, 6,127,175, 6,482,616, and 7,195,896, and PCT Publication Nos. WO1994/028152, WO1995/002697, WO1995/016772, WO1995/034671, WO1996/022378, WO1997/012986, WO1997/021826, and WO2003/022311.

The replication-deficient adenoviral vector of the invention can be produced in complementing cell lines that provide gene functions not present in the replication-deficient adenoviral vector, but required for viral propagation, at appropriate levels in order to generate high titers of viral vector stock. Such complementing cell lines are known and include, but are not limited to, 293 cells (described in, e.g., Graham et al. (1977) J. GEN. VIROL. 36: 59-72), PER.C6 cells (described in, e.g., PCT Publication No. WO1997/000326, and U.S. Pat. Nos. 5,994,128 and 6,033,908), and 293-ORF6 cells (described in, e.g., PCT Publication No. WO1995/034671 and Brough et al. (1997) J. VIROL. 71: 9206-9213). Other suitable complementing cell lines to produce the replication-deficient adenoviral vector of the invention include complementing cells that have been generated to propagate adenoviral vectors encoding transgenes whose expression inhibits viral growth in host cells (see, e.g., U.S. Patent Publication No. 2008/0233650). Additional suitable complementing cells are described in, for example, U.S. Pat. Nos. 6,677,156 and 6,682,929, and PCT Publication No. WO2003/020879. Formulations for adenoviral vector-containing compositions are further described in, for example, U.S. Pat. Nos. 6,225,289, and 6,514,943, and PCT Publication No. WO2000/034444.

Additional exemplary adenoviral vectors, and/or methods for making or propagating adenoviral vectors are described in U.S. Pat. Nos. 5,559,099, 5,837,511, 5,846,782, 5,851,806, 5,994,106, 5,994,128, 5,965,541, 5,981,225, 6,040,174, 6,020,191, 6,083,716, 6,113,913, 6,303,362, 7,067,310, and 9,073,980.

Commercially available adenoviral vector systems include the ViraPower™ Adenoviral Expression System available from Thermo Fisher Scientific, the AdEasy™ adenoviral vector system available from Agilent Technologies, and the Adeno-X™ Expression System 3 available from Takara Bio USA, Inc.

V. Host Cells and Cell Lines

Also encompassed by the invention are host cells or cell lines (e.g., prokaryotic or eukaryotic host cells or cell lines) that include a tRNA, aminoacyl-tRNA synthetase, unnatural amino acid, nucleic acid, and/or vector disclosed herein. The nucleic acid encoding the engineered tRNA and aminoacyl-tRNA synthetase can be expressed in an expression host cell either as an autonomously replicating vector within the expression host cell (e.g., a plasmid, or viral particle) or via a stable integrated element or series of stable integrated elements in the genome of the expression host cell, e.g., a mammalian host cell.

Host cells are genetically engineered (including but not limited to, transformed, transduced or transfected), for example, using nucleic acids or vectors disclosed herein. For example, in certain embodiments, one or more vectors include coding regions for an orthogonal tRNA, an orthogonal aminoacyl-tRNA synthetase, and, optionally, a protein to be modified by the inclusion of one or more UAAs, which are operably linked to gene expression control elements that are functional in the desired host cell or cell line. For example, the genes encoding tRNA synthetase and tRNA and an optional selectable marker (e.g., an antibiotic resistance gene, e.g., a puromycin resistance cassette) can be integrated in a transfer vector (e.g., a plasmid, which can be linearized prior to transfection), where for example, the genes encoding the tRNA synthetase can be under the control of a polymerase II promoter (e.g., CMV, EF1α, UbiC, or PGK, e.g., CMV or EF1α) and the genes encoding the tRNA can be under the control of a polymerase III promoter (e.g., U6, 7SK, or H1, e.g., U6). The vectors are transfected into cells and/or microorganisms by standard methods including electroporation or infection by viral vectors, and clones can selected via expression of the selectable marker (for example, by antibiotic resistance).

Exemplary prokaryotic host cells or cell lines include cells derived from a bacteria, e.g., Escherichia coli, Thermus thermophilus, Bacillus stearothermophilus, Pseudomonas fluorescens, Pseudomonas aeruginosa, and Pseudomonas putida. Exemplary eukaryotic host cells or cell lines include cells derived from a plant (e.g., a complex plant such as a monocot or dicot), an algae, a protist, a fungus, a yeast (including Saccharomyces cerevisiae), or an animal (including a mammal, an insect, an arthropod, etc.). Additional exemplary host cells or cell lines include HEK293, HEK293T, Expi293, CHO, CHOK1, Sf9, Sf21, HeLa, U20S, A549, HT1080, CAD, P19, NIH 3T3, L929, N2a, MCF-7, Y79, SO-RB50, HepG2, DUKX-X11, J558L, BHK, COS, Vero, NSO, or ESCs. It is understood that a host cell or cell line can include individual colonies, isolated populations (monoclonal), or a heterogeneous mixture of cells.

A contemplated cell or cell line includes, for example, one or multiple copies of an orthogonal tRNA/aminoacyl-tRNA synthetase pair, optionally stably maintained in the cell's genome or another piece of DNA maintained by the cell. For example, the cell or cell line may contain one or more copies of (i) a tryptophanyl tRNA/aminoacyl-tRNA synthetase pair (wild type or engineered) stably maintained by the cell, and/or (ii) a leucyl tRNA/aminoacyl-tRNA synthetase pair (wild-type or engineered) stably maintained by the cell.

For example, in certain embodiments, the cell line is a stable cell line and the cell line comprises a genome having stably integrated therein (i) a nucleic acid sequence encoding an aminoacyl-tRNA synthetase (e.g., a prokaryotic tryptophanyl-tRNA synthetase mutein capable of charging a tRNA with an unnatural amino acid or a prokaryotic leucyl-tRNA synthetase mutein capable of charging a tRNA with an unnatural amino acid, e.g., a tRNA synthetase mutein disclosed herein); and/or (ii) a nucleic acid sequence encoding a suppressor tRNA (e.g., prokaryotic suppressor tryptophanyl-tRNA capable of being charged with an unnatural amino acid or prokaryotic suppressor leucyl-tRNA capable of being charged with an unnatural amino acid, e.g., a suppressor tRNA disclosed herein).

Methods to introduce a nucleic acid encoding a tRNA and/or an aminoacyl-tRNA synthetase into the genome of a cell of interest, or to stably maintain the nucleic acid in DNA replicated by the cell that is outside of the genome, are well known in the art.

The nucleic acid encoding the tRNA and/or an aminoacyl-tRNA synthetase can be provided to the cell in an expression vector, transfer vector, or DNA cassette, e.g., an expression vector, transfer vector, or DNA cassette disclosed herein. The expression vector transfer vector, or DNA cassette encoding the tRNA and/or aminoacyl-tRNA synthetase can contain one or more copies of the tRNA and/or aminoacyl-tRNA synthetase optionally under the control of an inducible or constitutively active promoter. The expression vector, transfer vector, or DNA cassette may, for example, contain other standard components (enhancers, terminators, etc.). It is contemplated that the nucleic acid encoding the tRNA and the nucleic acid encoding the aminoacyl-tRNA synthetase may be on the same or different vector, may be present in the same or different ratios, and may be introduced into the cell, or stably integrated in the cellular genome, at the same time or sequentially.

One or multiple copies of a DNA cassette encoding the tRNA and/or aminoacyl-tRNA synthetase can be integrated into a host cell genome or stably maintained in the cell using a transposon system (e.g., PiggyBac), a viral vector (e.g., a lentiviral vector or other retroviral vector), CRISPR/Cas9 based recombination, electroporation and natural recombination, a BxB1 recombinase system, or using a replicating/maintained piece of DNA (such as one derived from Epstein-Barr virus).

In order to select for cell lines which stably maintain the nucleic acid encoding the tRNA and/or aminoacyl-tRNA synthetase and/or are efficient at incorporating UAAs into a protein of interest, a selectable marker can be used. Exemplary selectable markers include zeocin, puromycin, neomycin, dihydrofolate reductase (DHFR), glutamine synthetase (GS), mCherry-EGFP fusion, or other fluorescent proteins. In certain embodiments, a gene encoding a selectable marker protein (or a gene encoding a protein required for a detectable function, e.g., viability, in the presence of the selectable marker) may include a premature stop codon, such that the protein will only be expressed if the cell line is capable of incorporating a UAA at the site of the premature stop codon.

In certain embodiments, to develop a host cell or cell line including two or more tRNA/aminoacyl-tRNA synthetase pairs, one can use multiple identical or distinct UAA directing codons in order to identify host cells or cell lines which have incorporated multiple copies of the two or more tRNA/aminoacyl-tRNA synthetase pairs through iterative rounds of genomic integration and selection. Host cells or cell lines which contain enhanced UAA incorporation efficiency, low background, and decreased toxicity can first be isolated via a selectable marker containing one or more stop codons. Subsequently, the host cells or cell lines can be subjected to a selection scheme to identify host cells or cell lines which contain the desired copies of tRNA/aminoacyl-tRNA synthetase pairs and express a gene of interest (either genomically integrated or not) containing one or more stop codons. Protein expression may be assayed using any method known in the art, including for example, Western blot using an antibody that binds the protein of interest or a C-terminal tag.

The host cells or cell lines be cultured in conventional nutrient media modified as appropriate for such activities as, for example, screening steps, activating promoters or selecting transformants. These cells can optionally be cultured into transgenic organisms. Other useful references, e.g. for cell isolation and culture (e.g., for subsequent nucleic acid isolation) include Freshney (1994) Culture of Animal Cells, a Manual of Basic Technique, third edition, Wiley-Liss, New York; Payne et al. (1992) Plant Cell and Tissue Culture in Liquid Systems John Wiley & Sons, Inc. New York, N.Y.; Gamborg and Phillips (eds.) (1995) Plant Cell, Tissue and Organ Culture; Fundamental Methods Springer Lab Manual, Springer-Verlag (Berlin Heidelberg N.Y.) and Atlas and Parks (eds.) The Handbook of Microbiological Media (1993) CRC Press, Boca Raton, Fla.

The production of an exemplary cell line capable of producing antibodies incorporating a UAA is described in Roy et al. (2020) MABS 12(1), e1684749).

VI. Proteins Including Unnatural Amino Acids (UAAs) and Methods of Making the Same

Also encompassed by the invention are proteins including unnatural amino acids (UAAs) and methods of making the same.

The incorporation of an unnatural amino acid can be done for a variety of purposes, including tailoring changes in protein structure and/or function, changing size, acidity, nucleophilicity, hydrogen bonding, hydrophobicity, accessibility of protease target sites, targeting to a moiety (e.g., for a protein array), adding a biologically active molecule, attaching a polymer, attaching a radionuclide, modulating serum half-life, modulating tissue penetration (e.g. tumors), modulating active transport, modulating tissue, cell or organ specificity or distribution, modulating immunogenicity, modulating protease resistance, etc. Proteins that include an unnatural amino acid can have enhanced or even entirely new catalytic or biophysical properties. For example, the following properties are optionally modified by inclusion of an unnatural amino acid into a protein: toxicity, biodistribution, structural properties, spectroscopic properties, chemical and/or photochemical properties, catalytic ability, half-life (including but not limited to, serum half-life), ability to react with other molecules, including but not limited to, covalently or noncovalently, and the like. The compositions including proteins that include at least one unnatural amino acid are useful for, including but not limited to, novel therapeutics, diagnostics, enzymes, and binding proteins (e.g., therapeutic antibodies).

A protein may have at least one, for example, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least ten or more UAAs. The UAAs can be the same or different. For example, there can be 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more different sites in the protein that comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more different UAAs. A protein may have at least one, but fewer than all, of a particular amino acid present in the protein substituted with the UAA. For a given protein with more than one UAA, the UAA can be identical or different (for example, the protein can include two or more different types of UAAs, or can include two of the same UAA). For a given protein with more than two UAAs, the UAAs can be the same, different or a combination of a multiple unnatural amino acid of the same kind with at least one different UAA.

In certain embodiments, the protein is an antibody (or a fragment thereof, e.g., an antigen-binding fragment thereof), bispecific antibody, nanobody, affibody, viral protein, chemokine, cytokine, antigen, blood coagulation factor, hormone, growth factor, enzyme, cell signaling protein, or any other polypeptide or protein.

As used herein, unless otherwise indicated, the term “antibody” is understood to mean an intact antibody (e.g., an intact monoclonal antibody), or a fragment thereof, such as a Fc fragment of an antibody (e.g., an Fc fragment of a monoclonal antibody), or an antigen-binding fragment of an antibody (e.g., an antigen-binding fragment of a monoclonal antibody), including an intact antibody, antigen-binding fragment, or Fc fragment that has been modified, engineered, or chemically conjugated. Examples of antigen-binding fragments include Fab, Fab′, (Fab′)2, Fv, single chain antibodies (e.g., scFv), minibodies, and diabodies. Examples of antibodies that have been modified or engineered include chimeric antibodies, humanized antibodies, and multispecific antibodies (e.g., bispecific antibodies). An example of a chemically conjugated antibody is an antibody conjugated to a toxin moiety.

Typically, antibodies are multimeric proteins that contain four polypeptide chains. Two of the polypeptide chains are called immunoglobulin heavy chains (H chains), and two of the polypeptide chains are called immunoglobulin light chains (L chains). The immunoglobulin heavy and light chains are connected by an interchain disulfide bond. The immunoglobulin heavy chains are connected by interchain disulfide bonds. A light chain consists of one variable region (VL) and one constant region (CL). The heavy chain consists of one variable region (VH) and at least three constant regions (CH1, CH2 and CH3). The variable regions determine the binding specificity of the antibody.

The variable heavy (VH) and variable light (VL) regions can be further subdivided into regions of hypervariability, termed “complementarity determining regions” (“CDR”), interspersed with regions that are more conserved, termed “framework regions” (FR). Human antibodies have three VH CDRs and three VL CDRs, separated by framework regions FR1-FR4. The extent of the FRs and CDRs has been defined (Kabat, E. A., et al. (1991) SEQUENCES OF PROTEINS OF IMMUNOLOGICAL INTEREST, FIFTH EDITION, U.S. Department of Health and Human Services, NIH Publication No. 91-3242; and Chothia, C. et al. (1987) J. MOL. BIOL. 196:901-917). Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxyl-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.

An antibody molecule may have (i) a heavy chain constant region chosen from, e.g., the heavy chain constant regions of IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgD, and IgE; particularly, chosen from, e.g., the (e.g., human) heavy chain constant regions of IgG1, IgG2, IgG3, and IgG4, and/or (ii) a light chain constant region chosen from, e.g., the (e.g., human) light chain constant regions of kappa or lambda.

In certain embodiments, an UAA, e.g., a first and/or second UAA, is located in a heavy chain or light chain constant region of an antibody. For example, in certain embodiments, a protein comprising a first and/or second UAA is an antibody, or a fragment thereof, comprising a heavy chain constant region of human IgG1 isotype (for example, having the amino acid sequence of SEQ ID NO: 114, or an amino acid sequence having at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 114) and the first and/or second UAA is located within the heavy chain constant region of human IgG1 isotype (for example, at a position corresponding to the threonine at position 78 of SEQ ID NO: 114). In certain embodiments, a protein comprising a first and/or second UAA is an antibody, or a fragment thereof, comprising a human kappa constant region (for example, having the amino acid sequence of SEQ ID NO: 115, or an amino acid sequence having at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 115) and the first and/or second UAA is located within the human kappa constant region.

In certain embodiments, an UAA, e.g., a first and/or second UAA, is located in a framework region, e.g., FR4, of a variable heavy (VH) and/or a variable light (VL) region of an antibody.

In certain embodiments, a protein comprising a first and/or second UAA is an antibody, or a fragment thereof, comprising the amino acid sequence of SEQ ID NO: 116, or an amino acid sequence having at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 116, and the first and/or second UAA is located within SEQ ID NO: 116 (for example, at a position corresponding to the threonine at position 11 of SEQ ID NO: 116). In certain embodiments, a protein comprising a first and/or second UAA is an antibody, or a fragment thereof, comprising the amino acid sequence of SEQ ID NO: 117, or an amino acid sequence having at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 117, and the first and/or second UAA is located within SEQ ID NO: 117 (for example, at a position corresponding to the lysine at position 11 of SEQ ID NO: 117).

Additional examples of therapeutic, diagnostic, and other proteins that can be modified to comprise one or more unnatural amino acids are described in U.S. Patent Application Publication Nos. 2003/0082575 and 2005/0009049.

tRNAS, aminoacyl-tRNA synthetases, and/or unnatural amino acids disclosed herein may be used to incorporate an unnatural amino acid into a protein of interest using any appropriate translation system.

The term “translation system” refers to a system including components necessary to incorporate an amino acid into a growing polypeptide chain (protein). Components of a translation system can include, e.g., ribosomes, tRNA's, synthetases, mRNA and the like. Translation systems may be cellular or cell-free, and may be prokaryotic or eukaryotic. For example, translation systems may include, or be derived from, a non-eukaryotic cell, e.g., a bacterium (such as E. coli), a eukaryotic cell, e.g., a yeast cell, a mammalian cell, a plant cell, an algae cell, a fungus cell, or an insect cell.

Translation systems include host cells or cell lines, e.g., host cells or cell lines contemplated herein. To express a polypeptide of interest with an unnatural amino acid in a host cell, one may clone a polynucleotide encoding the polypeptide into an expression vector that contains, for example, a promoter to direct transcription, a transcription/translation terminator, and if for a nucleic acid encoding a protein, a ribosome binding site for translational initiation.

Translation systems also include whole cell preparations such as permeabilized cells or cell cultures wherein a desired nucleic acid sequence can be transcribed to mRNA and the mRNA translated. Cell-free translation systems are commercially available and many different types and systems are well-known. Examples of cell-free systems include, but are not limited to, prokaryotic lysates such as Escherichia coli lysates, and eukaryotic lysates such as wheat germ extracts, insect cell lysates, rabbit reticulocyte lysates, rabbit oocyte lysates and human cell lysates. Reconstituted translation systems may also be used. Reconstituted translation systems may include mixtures of purified translation factors as well as combinations of lysates or lysates supplemented with purified translation factors such as initiation factor-1 (IF-1), IF-2, IF-3 (α or β), elongation factor T (EF-Tu), or termination factors. Cell-free systems may also be coupled transcription/translation systems wherein DNA is introduced to the system, transcribed into mRNA and the mRNA is translated.

The invention provides methods of expressing a protein containing an unnatural amino acid and methods of producing a protein with one, or more, unnatural amino acids at specified positions in the protein. The methods comprise incubating a translation system (e.g., culturing or growing a host cell or cell line, e.g., a host cell or cell line disclosed herein) under conditions that permit incorporation of the unnatural amino acid into the protein being expressed in the cell. The translation system may be contacted with (e.g. the cell culture medium may be contacted with) one, or more, unnatural amino acids (e.g., leucyl or tryptophanyl analogs) under conditions suitable for incorporation of the one, or more, unnatural amino acids into the protein.

In certain embodiments, the protein is expressed from a nucleic acid sequence comprising a premature stop codon. The translation system (e.g., host cell or cell line) may, for example, contain a leucyl-tRNA synthetase mutein (e.g., a leucyl-tRNA synthetase mutein disclosed herein) capable of charging a suppressor leucyl tRNA (e.g., a suppressor leucyl tRNA disclosed herein) with an unnatural amino acid (e.g., a leucyl analog) which is incorporated into the protein at a position corresponding to the premature stop codon. In certain embodiments, the leucyl suppressor tRNA comprises an anticodon sequence that hybridizes to the premature stop codon and permits the unnatural amino to be incorporated into the protein at the position corresponding to the premature stop codon.

In certain embodiments, the protein is expressed from a nucleic acid sequence comprising a premature stop codon. The translation system (e.g., host cell or cell line) may, for example, contain a tryptophanyl-tRNA synthetase mutein (e.g., a tryptophanyl-tRNA synthetase mutein disclosed herein) capable of charging a suppressor tryptophanyl tRNA (e.g., a suppressor tryptophanyl tRNA disclosed herein) with an unnatural amino acid (e.g., a tryptophan analog) which is incorporated into the protein at a position corresponding to the premature stop codon. In certain embodiments, the tryptophanyl suppressor tRNA comprises an anticodon sequence that hybridizes to the premature stop codon and permits the unnatural amino to be incorporated into the protein at the position corresponding to the premature stop codon.

In certain embodiments, a protein (e.g., a protein containing a UAA, e.g., a first and/or second UAA) is expressed or produced in a eukaryotic cell (e.g., a mammalian cell). Features may distinguish proteins produced in prokaryotic cells (e.g., bacteria) from those produced in eukaryotic cells (e.g., mammalian cells). For example, proteins produced in mammalian cells may undergo post-translational modifications, e.g., modifications that are dependent upon enzymes located in organelles, e.g., the endoplasmic reticulum or Golgi apparatus. For example, disulfide bond formation in the endoplasmic reticulum may influence protein conformation and/or stabilization. Additional examples of such post-translational modifications include, without limitation, sulfation, amidation, palmitation, and glycosylation (e.g., N-linked glycosylation and O-linked glycosylation). Accordingly, in certain embodiments, a protein (e.g., a protein containing a UAA, e.g., a first and/or second UAA) comprises one or more post-translational modifications selected from sulfation, amidation, palmitation, and glycosylation (e.g., N-linked glycosylation and O-linked glycosylation).

Throughout the description, where compositions are described as having, including, or comprising specific components, or where processes and methods are described as having, including, or comprising specific steps, it is contemplated that, additionally, there are compositions of the present invention that consist essentially of, or consist of, the recited components, and that there are processes and methods according to the present invention that consist essentially of, or consist of, the recited processing steps.

In the application, where an element or component is said to be included in and/or selected from a list of recited elements or components, it should be understood that the element or component can be any one of the recited elements or components, or the element or component can be selected from a group consisting of two or more of the recited elements or components.

Further, it should be understood that elements and/or features of a composition or a method described herein can be combined in a variety of ways without departing from the spirit and scope of the present invention, whether explicit or implicit herein. For example, where reference is made to a particular compound, that compound can be used in various embodiments of compositions of the present invention and/or in methods of the present invention, unless otherwise understood from the context. In other words, within this application, embodiments have been described and depicted in a way that enables a clear and concise application to be written and drawn, but it is intended and will be appreciated that embodiments may be variously combined or separated without parting from the present teachings and invention(s). For example, it will be appreciated that all features described and depicted herein can be applicable to all aspects of the invention(s) described and depicted herein.

It should be understood that the expression “at least one of” includes individually each of the recited objects after the expression and the various combinations of two or more of the recited objects unless otherwise understood from the context and use. The expression “and/or” in connection with three or more recited objects should be understood to have the same meaning unless otherwise understood from the context.

The use of the term “include,” “includes,” “including,” “have,” “has,” “having,” “contain,” “contains,” or “containing,” including grammatical equivalents thereof, should be understood generally as open-ended and non-limiting, for example, not excluding additional unrecited elements or steps, unless otherwise specifically stated or understood from the context.

Where the use of the term “about” is before a quantitative value, the present invention also includes the specific quantitative value itself, unless specifically stated otherwise. As used herein, the term “about” refers to a 10% variation from the nominal value unless otherwise indicated or inferred.

It should be understood that the order of steps or order for performing certain actions is immaterial so long as the present invention remain operable. Moreover, two or more steps or actions may be conducted simultaneously.

The use of any and all examples, or exemplary language herein, for example, “such as” or “including,” is intended merely to illustrate better the present invention and does not pose a limitation on the scope of the invention unless claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the present invention.

SEQUENCE LISTING

SEQUENCE LISTING SEQ ID NO Description Sequence 1 E. coli MQEQYRPEEIESKVQLHWDEKRTFEVTEDESKEKYYCLSMLPYPSGR LeuRS LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDTFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWYYARYTCPQYKEGMLDSEA ANYWLPVDIYIGGIEHAIMHLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASPADMTLEW QESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDV HKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQE ALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDS TLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRK VIYVPGKLLNLVVGGPV 2 LeuRS.v1 MEEQYRPEEIESKVQLHWDKKRTFEVTEDESKEKYYCLSISPYPSGR LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDAFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWIYARYTCPQYKEGMLDSEA ANYWLPVDIAIGGIEHAIMGLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASPADMTLEW QESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDV HKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQE ALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDS TLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRK VIYVPGKLLNLVVGGPV 3 LeuRS.v2 MEEQYRPEEIESKVQLHWDKKRTFEVTEDESKEKYYCLSVSPYPSGR LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDRFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWSYARYTCPQYKEGMLDSEA ANYWLPVDILIGGIEHAIMGLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASPADMTLEW QESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDV HKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQE ALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDS TLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRK VIYVPGKLLNLVVGGPV 4 LeuRS.v3 MEEQYRPEEIESKVQLHWDEKRTFEVTEDESKEKYYCLSILPYPSGR LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDAFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWIYARYTCPQYKEGMLDSEA ANYWLPVDIAIGGIEHAIMGLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASPADMTLEW QESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDV HKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQE ALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDS TLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRK VIYVPGKLLNLVVGGPV 5 V9 MEEQYRPEEIESKVQLHWDMKRTFEVTEDESKEKYYCLSISPYPSGR LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDAFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWIYARYTCPQYKEGMLDSEA ANYWLPVDIAIGGIEHAIMGLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASPADMTLEW QESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDV HKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQE ALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDS TLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRK VIYVPGKLLNLVVGGPV 6 V10 MEEQYRPEEIESKVQLHWDVKRTFEVTEDESKEKYYCLSISPYPSGR LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDAFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWIYARYTCPQYKEGMLDSEA ANYWLPVDIAIGGIEHAIMGLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASPADMTLEW QESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDV HKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQE ALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDS TLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRK VIYVPGKLLNLVVGGPV 7 V26 MEEQYRPEEIESKVQLHWDKKRTFEVTEDESKEKYYCLSIVPYPSGR LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDAFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWIYARYTCPQYKEGMLDSEA ANYWLPVDIAIGGIEHAIMGLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASPADMTLEW QESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDV HKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQE ALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDS TLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRK VIYVPGKLLNLVVGGPV 8 V29 MEEQYRPEEIESKVQLHWDKKRTFEVTEDESKEKYYCLSIAPYPSGR LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDAFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWIYARYTCPQYKEGMLDSEA ANYWLPVDIAIGGIEHAIMGLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASPADMTLEW QESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDV HKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQE ALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDS TLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRK VIYVPGKLLNLVVGGPV 9 V48 MEEQYRPEEIESKVQLHWDkKRTFEVTEDESKEKYYCLSISPYPSGR LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDAFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWAYARYTCPQYKEGMLDSEA ANYWLPVDIAIGGIEHAIMGLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASPADMTLEW QESGVEGANRFKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDVH KTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQEA LLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDST LVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRKV IYVPGKLLNLVVGGPV 10 V50 MEEQYRPEEIESKVQLHWDkKRTFEVTEDESKEKYYCLSISPYPSGR LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDAFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWHYARYTCPQYKEGMLDSEA ANYWLPVDIAIGGIEHAIMGLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLEMMFASPADMTLEW QESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDV HKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQE ALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDS TLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRK VIYVPGKLLNLVVGGPV 11 V54 MEEQYRPEEIESKVQLHWDkKRTFEVTEDESKEKYYCLSISPYPSGR LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDAFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWIYARYTCPQYKEGMLDSEA ANYWLPVDIIIGGIEHAIMGLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASPADMTLEW QESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDV HKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQE ALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDS TLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRK VIYVPGKLLNLVVGGPV 12 V55 MEEQYRPEEIESKVQLHWDKKRTFEVTEDESKEKYYCLSISPYPSGR LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDAFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWIYARYTCPQYKEGMLDSEA ANYWLPVDIVIGGIEHAIMGLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASPADMTLEW QESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDV HKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQE ALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDS TLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRK VIYVPGKLLNLVVGGPV 13 V56 MEEQYRPEEIESKVQLHWDKKRTFEVTEDESKEKYYCLSISPYPSGR LHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAVKNN TAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQKFFT ELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVERKEI PQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEITFN VNDYDNTLTVYTTRPDAFMGCTYLAVAAGHPLAQKAAENNPELAAFI DECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFVLME YGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQALT EKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDWGVS RQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPIKAD PEWAKTTVNGMPALRETDTFDTFMESSWIYARYTCPQYKEGMLDSEA ANYWLPVDIGIGGIEHAIMGLLYFRFFHKLMRDAGMVNSDEPAKQLL CQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKAKDAAGHE LVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASPADMTLEW QESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQKALRRDV HKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQDRALMQE ALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADEKAMVEDS TLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYLDGVTVRK VIYVPGKLLNLVVGGPV 14 MX2EQYRPEEIESKVQLHWDX20KRTFEVTEDESKEKYYCLSX40X41P YPSGRLHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGA AVKNNTAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWE QKFFTELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKV ERKEIPQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGV EITFNVNDYDNTLTVYTTRPDX252FMGCTYLAVAAGHPLAQKAAENN PELAAFIDECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWA ANFVLMEYGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPD LSQQALTEKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYR LRDWGVSRQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGI TSPIKADPEWAKTTVNGMPALRETDTFDTFMESSWX499YARYTCPQY KEGMLDSEAANYWLPVDIX527IGGIEHAIMX537LLYFRFFHKLMRDA GMVNSDEPAKQLLCQGMVLADAFYYVGENGERNWVSPVDAIVERDEK GRIVKAKDAAGHELVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLF MMFASPADMTLEWQESGVEGANRFLKRVWKLVYEHTAKGDVAALNVD ALTENQKALRRDVHKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKA PTDGEQDRALMQEALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAP WPVADEKAMVEDSTLVVVQVNGKVRAKITVPVDATEEQVRERAGQEH LVAKYLDGVTVRKVIYVPGKLLNLVVGGPV wherein X2 is Q or E, X20 is E, K, V or M, X40 is M, I, or V, X41 is L, S, V, or A, X252 is T, A, or R, X499 is Y, A, I, H, or S, X527 is Y, A, I, L, or V, and X537 is H or G 15 MEEQYRPEEIESKVQLHWDX20KRTFEVTEDESKEKYYCLSIX41PYP SGRLHMGHVRNYTIGDVIARYQRMLGKNVLQPIGWDAFGLPAEGAAV KNNTAPAPWTYDNIAYMKNQLKMLGFGYDWSRELATCTPEYYRWEQK FFTELYKKGLVYKKTSAVNWCPNDQTVLANEQVIDGCCWRCDTKVER KEIPQWFIKITAYADELLNDLDKLDHWPDTVKTMQRNWIGRSEGVEI TFNVNDYDNTLTVYTTRPDAFMGCTYLAVAAGHPLAQKAAENNPELA AFIDECRNTKVAEAEMATMEKKGVDTGFKAVHPLTGEEIPVWAANFV LMEYGTGAVMAVPGHDQRDYEFASKYGLNIKPVILAADGSEPDLSQQ ALTEKGVLFNSGEFNGLDHEAAFNAIADKLTAMGVGERKVNYRLRDW GVSRQRYWGAPIPMVTLEDGTVMPTPDDQLPVILPEDVVMDGITSPI KADPEWAKTTVNGMPALRETDTFDTFMESSWX499YARYTCPQYKEGM LDSEAANYWLPVDIX527IGGIEHAIMGLLYFRFFHKLMRDAGMVNSD EPAKQLLCQGMVLADAFYYVGENGERNWVSPVDAIVERDEKGRIVKA KDAAGHELVYTGMSKMSKSKNNGIDPQVMVERYGADTVRLFMMFASP ADMTLEWQESGVEGANRFLKRVWKLVYEHTAKGDVAALNVDALTENQ KALRRDVHKTIAKVTDDIGRRQTFNTAIAAIMELMNKLAKAPTDGEQ DRALMQEALLAVVRMLNPFTPHICFTLWQELKGEGDIDNAPWPVADE KAMVEDSTLVVVQVNGKVRAKITVPVDATEEQVRERAGQEHLVAKYL DGVTVRKVIYVPGKLLNLVVGGPV wherein X20 is K, V or M, X41 is S, V, or A, X499 is A, I,or H, and X527 is A, I, or V 16 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGATTctaAATCCCTCGG tRNA. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGtA wtCUA 17 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGATTtcaAATCCCTCGG tRNA. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGtA wtUCA 18 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGATTttaAATCCCTCGG tRNA. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGtA wtUUA 19 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGACTctaAATCCCTCGG tRNA. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGcA h1CUA 20 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGACTtcaAATCCCTCGG tRNA. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGcA h1UCA 21 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGACTttaAATCCCTCGG tRNA. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGcA h1UUA 22 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGATTctaAATCCCTCGG tRNA. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGtACCA wtCUA_cca 23 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGATTtcaAATCCCTCGG tRNA. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGtACCA wtUCA_cca 24 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGATTttaAATCCCTCGG tRNA. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGtACCA wtUUA_cca 25 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGACTctaAATCCCTCGG tRNA. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGcACCA h1CUA_cca 26 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGACTtcaAATCCCTCGG tRNA. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGcACCA h1UCA_cca 27 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGACTttaAATCCCTCGG tRNA. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGcACCA h1UUA_cca 28 Leu- GCCCGGATGGTGGAATCGGTAGACACAAGGGACTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCCGGGCACCA 30 wherein nnn is CTA, TCA, or TTA 29 Leu- GGGCGTGTGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCCGCGCCCACCA 31 wherein nnn is CTA, TCA, or TTA 30 Leu- GGGCGCGTGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCCGCGCCCACCA 32 wherein nnn is CTA, TCA, or TTA 31 Leu- GGGCATGTGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCCATGCCCACCA 33 wherein nnn is CTA, TCA, or TTA 32 Leu- GGGCACGTGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCCGTGCCCACCA 34 wherein nnn is CTA, TCA, or TTA 33 Leu- GGGGGTGTGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCCGCCCCCACCA 35 wherein nnn is CTA, TCA, or TTA 34 Leu- GGGGGCGTGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCCGTCCCCACCA 36 wherein nnn is CTA, TCA, or TTA 35 Leu- GGGGATGTGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCCGTCCCCACCA 37 wherein nnn is CTA, TCA, or TTA 36 Leu- GGGGACGTGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCCGTCCCCACCA 38 wherein nnn is CTA, TCA, or TTA 37 Leu- GCCCGTATGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTGCGGGCACCA 39 wherein nnn is CTA, TCA, or TTA 38 Leu- GGGATAGTGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCCTATCCCACCA 40 wherein nnn is CTA, TCA, or TTA 39 Leu- GGGCATGTGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCCGTGCCCACCA 41 wherein nnn is CTA, TCA, or TTA 40 Leu- GGGCAGATGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTCTGCCCACCA 42 wherein nnn is CTA, TCA, or TTA 41 Leu- GGGCGTATGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTGCGCCCACCA 43 wherein nnn is CTA, TCA, or TTA 42 Leu- GGGCAAGTGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCCTTGCCCACCA 44 wherein nnn is CTA, TCA, or TTA 43 Leu- GCACACATGGTGGAATCGGTAGACACAAGGGATTnnnAATCCCTCGG tRNA.003. CGTTCGCGCTGTGCGGGTTCAAGTCCCGCTGTGTGCACCA 45 wherein nnn is CTA, TCA, or TTA 44 E. coli MTKPIVFSGAQPSGELTIGNYMGALRQWVNMQDDYHCIYCIVDQHAI TrpRS.wt TVRQDAQKLRKATLDTLALYLACGIDPEKSTIFVQSHVPEHAQLGWA LNCYTYFGELSRMTQFKDKSARYAENINAGLFDYPVLMAADILLYQT NLVPVGEDQKQHLELSRDIAQRFNALYGEIFKVPEPFIPKSGARVMS LLEPTKKMSKSDDNRNNVIGLLEDPKSVVKKIKRAVTDSDEPPVVRY DVQNKAGVSNLLDILSAVTGQSIPELEKQFEGKMYGHLKGEVADAVS GMLTELQERYHRFRNDEAFLQQVMKDGAEKASAHASRTLKAVYEAIG FVAKP 45 TrpRS.h14 MTKPIVFAGAQPSGELTIGNYMGALRQWVNMQDDYHCIYCIVDQHAI TVRQDAQKLRKATLDTLALYLACGIDPEKSTIFVQSHVPEHAQLGWA LNCYTYFGELSRMTQFKDKSARYAENINAGLFDYPVLMAADILLYQT NLGPCGEDQKQHLELSRDIAQRFNALYGEIFKVPEPFIPKSGARVMS LLEPTKKMSKSDDNRNNVIGLLEDPKSVVKKIKRAVTDSDEPPVVRY DVQNKAGVSNLLDILSAVTGQSIPELEKQFEGKMYGHLKGEVADAVS GMLTELQERYHRFRNDEAFLQQVMKDGAEKASAHASRTLKAVYEAIG FVAKP 46 TrpRS.h13 MTKPIVFAGAQPSGELTIGNYMGALRQWVNMQDDYHCIYCIVDQHAI TVRQDAQKLRKATLDTLALYLACGIDPEKSTIFVQSHVPEHAQLGWA LNCYTYFGELSRMTQFKDKSARYAENINAGLFDYPVLMAADILLYQT NLAPAGEDQKQHLELSRDIAQRFNALYGEIFKVPEPFIPKSGARVMS LLEPTKKMSKSDDNRNNVIGLLEDPKSVVKKIKRAVTDSDEPPVVRY DVQNKAGVSNLLDILSAVTGQSIPELEKQFEGKMYGHLKGEVADAVS GMLTELQERYHRFRNDEAFLQQVMKDGAEKASAHASRTLKAVYEAIG FVAKP 47 TrpRS.h9 MTKPIVFAGAQPSGELTIGNYMGALRQWVNMQDDYHCIYCIVDQHAI TVRQDAQKLRKATLDTLALYLACGIDPEKSTIFVQSHVPEHAQLGWA LNCYTYFGELSRMTQFKDKSARYAENINAGLFDYPVLMAADILLYQT NLSPAGEDQKQHLELSRDIAQRFNALYGEIFKVPEPFIPKSGARVMS LLEPTKKMSKSDDNRNNVIGLLEDPKSVVKKIKRAVTDSDEPPVVRY DVQNKAGVSNLLDILSAVTGQSIPELEKQFEGKMYGHLKGEVADAVS GMLTELQERYHRFRNDEAFLQQVMKDGAEKASAHASRTLKAVYEAIG FVAKP 48 TrpRS.h10 MTKPIVFAGAQPSGELTIGNYMGALRQWVNMQDDYHCIYCIVDQHAI TVRQDAQKLRKATLDTLALYLACGIDPEKSTIFVQSHVPEHAQLGWA LNCYTYFGELSRMTQFKDKSARYAENINAGLFDYPVLMAADILLYQT NLGPAGEDQKQHLELSRDIAQRFNALYGEIFKVPEPFIPKSGARVMS LLEPTKKMSKSDDNRNNVIGLLEDPKSVVKKIKRAVTDSDEPPVVRY DVQNKAGVSNLLDILSAVTGQSIPELEKQFEGKMYGHLKGEVADAVS GMLTELQERYHRFRNDEAFLQQVMKDGAEKASAHASRTLKAVYEAIG FVAKP 49 E. coli AGGGGCGTAGTTCAATTGGTAGAGCACCGGTCTccaAAACCGGGTGT Trp-tRNA TGGGAGTTCGAGTCTCTCCGCCCCTGCCA 50 Trp- AGGGGCGTAGTTCAATTGGTAGAGCACCGGTCTctaAAACCGGGTGT tRNACUA TGGGAGTTCGAGTCTCTCCGCCCCTGCCA 51 Trp- AGGGGCGTAGTTCAATTGGTAGAGCACCGGTCTtcaAAACCGGGTGT tRNAUCA TGGGAGTTCGAGTCTCTCCGCCCCTGCCA 52 Trp- AGGGGCGTAGTTCAATTGGTAGAGCACCGGTCTttaAAACCGGGTGT tRNAUUA TGGGAGTTCGAGTCTCTCCGCCCCTGCCA 53 Trp- TGGGGTATCGCCAAGCGGTAAGGCACCGGATTCnnnTTCCGGCATTC tRNA.001.4 CGAGGTTCGAATCCTCGTACCCCAGCCA wherein nnn is CTA, TCA, or TTA 54 Trp- AGGGGCATAGCTCAAGCGGTAAAGCACCGGACTnnnAAACCGGCAGT tRNA.001.5 CCGAAGTTCGAATCCCCCCACCCCAGCCA wherein nnn is CTA, TCA, or TTA 55 E. coli ATGCAGGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT LeuRS GCATTGGGATGAAAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCATGCTGCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TACCTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGTATTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTTATATTGGCGGCATTGAACA TGCGATTATGCATCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTCTGAAACGCGTGTG GAAACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGA ACGTGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTG CATAAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGAC CTTTAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGG CGAAAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAA GCGCTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATAT TTGCTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATA ACGCGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGC ACCCTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTAC CGTGCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCC AGGAACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAA GTGATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCC GGTG 56 LeuRS.v1 ATGGAAGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT GCATTGGGATAAAAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCATTAGCCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TGCGTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGATTTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTGCGATTGGCGGCATTGAACA TGCGATTATGGGCCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTCTGAAACGCGTGTG GAAACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGA ACGTGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTG CATAAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGAC CTTTAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGG CGAAAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAA GCGCTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATAT TTGCTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATA ACGCGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGC ACCCTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTAC CGTGCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCC AGGAACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAA GTGATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCC GGTG 57 LeuRS.v2 ATGGAAGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT GCATTGGGATAAAAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCGTGAGCCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TCGCTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGAGCTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTCTGATTGGCGGCATTGAACA TGCGATTATGGGCCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTCTGAAACGCGTGTG GAAACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGA ACGTGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTG CATAAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGAC CTTTAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGG CGAAAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAA GCGCTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATAT TTGCTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATA ACGCGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGC ACCCTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTAC CGTGCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCC AGGAACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAA GTGATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCC GGTG 58 LeuRS.v3 ATGGAAGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT GCATTGGGATGAAAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCATTCTGCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TGCGTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGATTTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTGCGATTGGCGGCATTGAACA TGCGATTATGGGCCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTCTGAAACGCGTGTG GAAACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGA ACGTGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTG CATAAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGAC CTTTAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGG CGAAAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAA GCGCTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATAT TTGCTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATA ACGCGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGC ACCCTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTAC CGTGCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCC AGGAACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAA GTGATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCC GGTG 59 V9 ATGGAAGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT GCATTGGGATATGAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCATTAGCCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TGCGTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGATTTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTGCGATTGGCGGCATTGAACA TGCGATTATGGGCCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTCTGAAACGCGTGTG GAAACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGA ACGTGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTG CATAAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGAC CTTTAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGG CGAAAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAA GCGCTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATAT TTGCTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATA ACGCGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGC ACCCTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTAC CGTGCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCC AGGAACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAA GTGATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCC GGTG 60 V10 ATGGAAGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT GCATTGGGATGTGAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCATTAGCCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TGCGTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGATTTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTGCGATTGGCGGCATTGAACA TGCGATTATGGGCCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTCTGAAACGCGTGTG GAAACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGA ACGTGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTG CATAAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGAC CTTTAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGG CGAAAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAA GCGCTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATAT TTGCTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATA ACGCGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGC ACCCTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTAC CGTGCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCC AGGAACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAA GTGATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCC GGTG 61 V26 ATGGAAGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT GCATTGGGATAAAAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCATTGTGCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TGCGTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGATTTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTGCGATTGGCGGCATTGAACA TGCGATTATGGGCCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTCTGAAACGCGTGTG GAAACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGA ACGTGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTG CATAAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGAC CTTTAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGG CGAAAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAA GCGCTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATAT TTGCTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATA ACGCGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGC ACCCTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTAC CGTGCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCC AGGAACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAA GTGATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCC GGTG 62 V29 ATGGAAGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT GCATTGGGATAAAAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCATTGCGCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TGCGTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGATTTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTGCGATTGGCGGCATTGAACA TGCGATTATGGGCCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTCTGAAACGCGTGTG GAAACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGA ACGTGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTG CATAAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGAC CTTTAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGG CGAAAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAA GCGCTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATAT TTGCTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATA ACGCGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGC ACCCTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTAC CGTGCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCC AGGAACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAA GTGATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCC GGTG 63 V48 ATGGAAGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT GCATTGGGATAAAAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCATTAGCCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TGCGTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGGCGTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTGCGATTGGCGGCATTGAACA TGCGATTATGGGCCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTAAACGCGTGTGGAA ACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGAACG TGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTGCAT AAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGACCTT TAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGGCGA AAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAAGCG CTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATATTTG CTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATAACG CGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGCACC CTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTACCGT GCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCCAGG AACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAAGTG ATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCCGGT G 64 V50 ATGGAAGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT GCATTGGGATAAAAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCATTAGCCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TGCGTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGCATTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTGCGATTGGCGGCATTGAACA TGCGATTATGGGCCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTCTGAAACGCGTGTG GAAACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGA ACGTGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTG CATAAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGAC CTTTAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGG CGAAAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAA GCGCTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATAT TTGCTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATA ACGCGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGC ACCCTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTAC CGTGCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCC AGGAACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAA GTGATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCC GGTG 65 V54 ATGGAAGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT GCATTGGGATAAAAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCATTAGCCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TGCGTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGATTTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTATTATTGGCGGCATTGAACA TGCGATTATGGGCCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTCTGAAACGCGTGTG GAAACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGA ACGTGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTG CATAAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGAC CTTTAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGG CGAAAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAA GCGCTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATAT TTGCTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATA ACGCGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGC ACCCTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTAC CGTGCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCC AGGAACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAA GTGATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCC GGTG 66 V55 ATGGAAGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT GCATTGGGATAAAAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCATTAGCCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TGCGTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGATTTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTGTGATTGGCGGCATTGAACA TGCGATTATGGGCCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTCTGAAACGCGTGTG GAAACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGA ACGTGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTG CATAAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGAC CTTTAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGG CGAAAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAA GCGCTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATAT TTGCTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATA ACGCGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGC ACCCTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTAC CGTGCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCC AGGAACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAA GTGATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCC GGTG 67 V56 ATGGAAGAACAGTATCGCCCGGAAGAAATTGAAAGCAAAGTGCAGCT GCATTGGGATAAAAAACGCACCTTTGAAGTGACCGAAGATGAAAGCA AAGAAAAATATTATTGCCTGAGCATTAGCCCGTATCCGAGCGGCCGC CTGCATATGGGCCATGTGCGCAACTATACCATTGGCGATGTGATTGC GCGCTATCAGCGCATGCTGGGCAAAAACGTGCTGCAGCCGATTGGCT GGGATGCGTTTGGCCTGCCGGCGGAAGGCGCGGCGGTGAAAAACAAC ACCGCGCCGGCGCCGTGGACCTATGATAACATTGCGTATATGAAAAA CCAGCTGAAAATGCTGGGCTTTGGCTATGATTGGAGCCGCGAACTGG CGACCTGCACCCCGGAATATTATCGCTGGGAACAGAAATTTTTTACC GAACTGTATAAAAAAGGCCTGGTGTATAAAAAAACCAGCGCGGTGAA CTGGTGCCCGAACGATCAGACCGTGCTGGCGAACGAACAGGTGATTG ATGGCTGCTGCTGGCGCTGCGATACCAAAGTGGAACGCAAAGAAATT CCGCAGTGGTTTATTAAAATTACCGCGTATGCGGATGAACTGCTGAA CGATCTGGATAAACTGGATCATTGGCCGGATACCGTGAAAACCATGC AGCGCAACTGGATTGGCCGCAGCGAAGGCGTGGAAATTACCTTTAAC GTGAACGATTATGATAACACCCTGACCGTGTATACCACCCGCCCGGA TGCGTTTATGGGCTGCACCTATCTGGCGGTGGCGGCGGGCCATCCGC TGGCGCAGAAAGCGGCGGAAAACAACCCGGAACTGGCGGCGTTTATT GATGAATGCCGCAACACCAAAGTGGCGGAAGCGGAAATGGCGACCAT GGAAAAAAAAGGCGTGGATACCGGCTTTAAAGCGGTGCATCCGCTGA CCGGCGAAGAAATTCCGGTGTGGGCGGCGAACTTTGTGCTGATGGAA TATGGCACCGGCGCGGTGATGGCGGTGCCGGGCCATGATCAGCGCGA TTATGAATTTGCGAGCAAATATGGCCTGAACATTAAACCGGTGATTC TGGCGGCGGATGGCAGCGAACCGGATCTGAGCCAGCAGGCGCTGACC GAAAAAGGCGTGCTGTTTAACAGCGGCGAATTTAACGGCCTGGATCA TGAAGCGGCGTTTAACGCGATTGCGGATAAACTGACCGCGATGGGCG TGGGCGAACGCAAAGTGAACTATCGCCTGCGCGATTGGGGCGTGAGC CGCCAGCGCTATTGGGGCGCGCCGATTCCGATGGTGACCCTGGAAGA TGGCACCGTGATGCCGACCCCGGATGATCAGCTGCCGGTGATTCTGC CGGAAGATGTGGTGATGGATGGCATTACCAGCCCGATTAAAGCGGAT CCGGAATGGGCGAAAACCACCGTGAACGGCATGCCGGCGCTGCGCGA AACCGATACCTTTGATACCTTTATGGAAAGCAGCTGGATTTATGCGC GCTATACCTGCCCGCAGTATAAAGAAGGCATGCTGGATAGCGAAGCG GCGAACTATTGGCTGCCGGTGGATATTGGCATTGGCGGCATTGAACA TGCGATTATGGGCCTGCTGTATTTTCGCTTTTTTCATAAACTGATGC GCGATGCGGGCATGGTGAACAGCGATGAACCGGCGAAACAGCTGCTG TGCCAGGGCATGGTGCTGGCGGATGCGTTTTATTATGTGGGCGAAAA CGGCGAACGCAACTGGGTGAGCCCGGTGGATGCGATTGTGGAACGCG ATGAAAAAGGCCGCATTGTGAAAGCGAAAGATGCGGCGGGCCATGAA CTGGTGTATACCGGCATGAGCAAAATGAGCAAAAGCAAAAACAACGG CATTGATCCGCAGGTGATGGTGGAACGCTATGGCGCGGATACCGTGC GCCTGTTTATGATGTTTGCGAGCCCGGCGGATATGACCCTGGAATGG CAGGAAAGCGGCGTGGAAGGCGCGAACCGCTTTCTGAAACGCGTGTG GAAACTGGTGTATGAACATACCGCGAAAGGCGATGTGGCGGCGCTGA ACGTGGATGCGCTGACCGAAAACCAGAAAGCGCTGCGCCGCGATGTG CATAAAACCATTGCGAAAGTGACCGATGATATTGGCCGCCGCCAGAC CTTTAACACCGCGATTGCGGCGATTATGGAACTGATGAACAAACTGG CGAAAGCGCCGACCGATGGCGAACAGGATCGCGCGCTGATGCAGGAA GCGCTGCTGGCGGTGGTGCGCATGCTGAACCCGTTTACCCCGCATAT TTGCTTTACCCTGTGGCAGGAACTGAAAGGCGAAGGCGATATTGATA ACGCGCCGTGGCCGGTGGCGGATGAAAAAGCGATGGTGGAAGATAGC ACCCTGGTGGTGGTGCAGGTGAACGGCAAAGTGCGCGCGAAAATTAC CGTGCCGGTGGATGCGACCGAAGAACAGGTGCGCGAACGCGCGGGCC AGGAACATCTGGTGGCGAAATATCTGGATGGCGTGACCGTGCGCAAA GTGATTTATGTGCCGGGCAAACTGCTGAACCTGGTGGTGGGCGGCCC GGTG 68 GTGGGGTTCCCGAGCGGCCAAAGGGAGCAGACTtcAAATCTGCCGTC ACAGACTTCGAAGGTTCGAATCCTTCCCCCACCA 69 GTGGGGTTCCCGAGCGGCCAAAGGGAGCAGACTnnnAATCTGCCGTC ACAGACTTCGAAGGTTCGAATCCTTCCCCCACCA wherein nnn is CTA, TCA, or TTA 70 MASSNLIKQLQERGLVAQVTDEEALAERLAQGPIALVCGEDPTADSL HLGHLVPLLCLKRFQQAGHKPVALVGGATGLIGDPSFKAAERKLNTE ETVQEWVDKIRKQVAPFLDFDCGENSAIAANNYDWFGNMNVLTFLRD IGKHFSVNQMINKEAVKQRLNREDQGISFTEFSYNLLQGYSMACLNK QYGVVLQIGGSDQWGNITSGIDLTRRLHQNQVFGLTVPLITKADGTK FGKTEGGAVWLDPKKTSPYKFYQFWINTADADVYRFLKFFTFMSIEE INALEEEDKNSGKAPRAQYVLAEQVTRLVHGEEGLQAAKRITECLFS GSLSALSEADFEQLAQDGVPMVEMEKGADLMQALVDSELQPSRGQAR KTIASNAITINGEKQSDPEYFFKEEDRLFGRFTLLRRGKKNYCLICW KGPV 71 atggcaagcagtaacttgattaaacaattgcaagagcgggggctggt agcccaggtgacggacgaggaagcgttagcagagcgactggcgcaag gcccgatcgcactcGTGtgtggcttcgatcctaccgctgacagcttg catttggggcatcttgttccattgttatgcctgaaacgcttccagca ggcgggccacaagccggttgcgctggtaggcggcgcgacgggtctga ttggcgacccgagcttcaaagctgccgagcgtaagctgaacaccgaa gaaactgttcaggagtgggtggacaaaatccgtaagcaggttgcccc gttcctcgatttcgactgtggagaaaactctgctatcgcggccaata attatgactggttcggcaatatgaatgtgctgaccttcctgcgcgat attggcaaacacttctccgttaaccagatgatcaacaaagaagcggt taagcagcgtctcaaccgtgaagAtcaggggatttcgttcactgagt tttcctacaacctgctGcagggttatAgtatggcctgtTTGaacaaa cagtacggtgtggtgctgcaaattggtggttctgaccaatggggtaa catcacttctggtatcgacctgacccgtcgtctgcatcagaatcagg tgtttggcctgaccgttccgctgatcactaaagcagatggcaccaaa tttggtaaaactgaaggcggcgcagtctggttggacccgaagaaaac cagcccgtacaaattctaccagttctggatcaacactgcggatgccg acgtttaccgcttcctgaagttcttcacctttatgagcattgaagag atcaacgccctggaagaagaagataaaaacagcggtaaagcaccgcg cgcccagtatgtactggcggagcaggtgactcgtctggttcacggtg aagaaggtttacaggcggcaaaacgtattaccgaatgcctgttcagc ggttctttgagtgcgctgagtgaagcggacttcgaacagctggcgca ggacggcgtaccgatggttgagatggaaaagggcgcagacctgatgc aggcactggtcgattctgaactgcaaccttcccgtggtcaggcacgt aaaactatcgcctccaatgccatcaccattaacggtgaaaaacagtc cgatcctgaatacttctttaaagaagaagatcgtctgtttggtcgtt ttaccttactgcgtcgcggtaaaaagaattactgtctgatttgetgg aaagggcccgtttaa 72 GGAAACCTGATCATGTAGATCGAACGGACTCTAAATCCGTTCAGCCG GGTTAGATTCCCGGGGTTTCCG 73 GGAAACCTGATCATGTAGATCGAACGGACTnnnAATCCGTTCAGCCG GGTTAGATTCCCGGGGTTTCCG 74 gGAAACCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGUUUCcgcca 75 gGGCGGCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGCUGCCcgcca 76 gGGUGACugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGUUGCCcgcca 77 gGGGGGCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGCUGCCcgcca 78 gGGCGGCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGUUGCCcgcca 79 gGGCGCCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGCGCCcgcca 80 gGGGAGGugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggCCUCCCcgcca 81 gGGGACCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGUCCCcgcca 82 gGCCGGGugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggCCUGGCcgcca 83 gGGGACCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGUCCUcgcca 84 gGGGCCCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGGUCCcgcca 85 gGGGGCCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGGUCCcgcca 86 gGGGUCCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGAUCCcgcca 87 gGGGACCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGUUCCcgcca 88 gGGGAGGugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggUCUUCCcgcca 89 gGGGGGGugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggCCCCCUcgcca 90 gGUGGGGugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggCCCUACcgcca 91 gGGGGUCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGACUCCcgcca 92 gGGUCCCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGGGUCcgcca 93 gGGGACCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGUUCUcgcca 94 gGGCGGCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGCCGCCcgcca 95 gAGCACCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGUGCUcgcca 96 gGGGGGGugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggCCCCCCcgcca 97 gAGGGGGugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggCCCCCUcgcca 98 gGGAGCCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGUUCCcgcca 99 gGGAGCCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGUUCCcgcca 100 gAGGACCugaucauguagaucgaacggacucuaaauccguucagccg gguuagauucccggGGUUCUcgcca 101 MDKKPLDVLISATGLWMSRTGTLHKIKHHEVSRSKIYIEMACGDHLV VNNSRSCRTARAFRHHKYRKTCKRCRVSDEDINNFLTRSTESKNSVK VRVVSAPKVKKAMPKSVSRAPKPLENSVSAKASTNTSRSVPSPAKST PNSSVPASAPAPSLTRSQLDRVEALLSPEDKISLNMAKPFRELEPEL VTRRKNDFQRLYTNDREDYLGKLERDITKFFVDRGFLEIKSPILIPA EYVERMGINNDTELSKQIFRVDKNLCLRPMLAPTLYNYLRKLDRILP GPIKIFEVGPCYRKESDGKEHLEEFTMVNFCQMGSGCTRENLEALIK EFLDYLEIDFEIVGDSCMVFGDTLDIMHGDLELSSAVVGPVSLDREW GIDKPWIGAGFGLERLLKVMHGFKNIKRASRSESYYNGISTNL 102 ATGGATAAAAAACCATTAGATGTTTTAATATCTGCGACCGGGCTCTG GATGTCCAGGACTGGCACGCTCCACAAAATCAAGCACCATGAGGTCT CAAGAAGTAAAATATACATTGAAATGGCGTGTGGAGACCATCTTGTT GTGAATAATTCCAGGAGTTGTAGAACAGCCAGAGCATTCAGACATCA TAAGTACAGAAAAACCTGCAAACGATGTAGGGTTTCGGACGAGGATA TCAATAATTTTCTCACAAGATCAACCGAAAGCAAAAACAGTGTGAAA GTTAGGGTAGTTTCTGCTCCAAAGGTCAAAAAAGCTATGCCGAAATC AGTTTCAAGGGCTCCGAAGCCTCTGGAAAATTCTGTTTCTGCAAAGG CATCAACGAACACATCCAGATCTGTACCTTCGCCTGCAAAATCAACT CCAAATTCGTCTGTTCCCGCATCGGCTCCTGCTCCTTCACTTACAAG AAGCCAGCTTGATAGGGTTGAGGCTCTCTTAAGTCCAGAGGATAAAA TTTCTCTAAATATGGCAAAGCCTTTCAGGGAACTTGAGCCTGAACTT GTGACAAGAAGAAAAAACGATTTTCAGCGGCTCTATACCAATGATAG AGAAGACTAGCTCGGTAAACTCGAACGTGATATTACGAAATTTTTCG TAGACCGGGGTTTTCTGGAGATAAAGTCTCCTATCCTTATTCCGGCG GAATACGTGGAGAGAATGGGTATTAATAATGATACTGAACTTTCAAA ACAGATCTTCCGGGTGGATAAAAATCTCTGCTTGAGGCCAATGCTTG CCCCGACTCTTTACAACTATCTGCGAAAACTCGATAGGATTTTACCA GGCCCAATAAAAATTTTCGAAGTCGGACCTTGTTACCGGAAAGAGTC TGACGGCAAAGAGCACCTGGAAGAATTTAGTATGGTGAACTTCTGTC AGATGGGTTCGGGATGTACTCGGGAAAATCTTGAAGCTCTCATCAAA GAGTTTCTGGACTATCTGGAAATCGACTTCGAAATCGTAGGAGATTC CTGTATGGTCTtTGGGGATACTCTTGATATAATGCACGGGGACCTGG AGCTTTCTTCGGCAGTCGTCGGGCCAGTTTCTCTTGATAGAGAATGG GGTATTGACAAACCATGGATAGGTGCAGGTTTTGGTCTTGAACGCTT GCTCAAGGTTATGCACGGCTTTAAAAACATTAAGAGGGCATCAAGGT CCGAATCTTACTATAATGGGATTTCAACCAATCTGTAA 103 TrpRS.h14 ATGACTAAGCCCATCGTTTTTgctGGCGCACAGCCCTCAGGTGAATT GACCATTGGTAACTACATGGGTGCGCTGCGTCAGTGGGTAAACATGC AGGATGACTACCATTGCATTTACTGTATCGTTGACCAACACGCGATC ACCGTGCGCCAGGATGCACAGAAGCTGCGTAAAGCGACGCTGGATAC GCTGGCCTTGTATCTGGCTTGTGGTATCGATCCTGAGAAAAGCACCA TTTTTGTTCAGTCCCACGTGCCGGAACATGCACAGTTAGGCTGGGCA CTGAACTGCTATACCTACTTCGGCGAACTGAGTCGCATGACGCAGTT TAAAGATAAATCTGCGCGTTATGCCGAGAACATCAACGCTGGTCTGT TTGACTATCCGGTGCTGATGGCAGCGGACATCCTGCTGTATCAAACT AATCTGggtccttgtGGTGAAGACCAGAAACAGCACCTCGAACTGAG CCGCGATATTGCCCAGCGTTTCAACGCGCTGTATGGCGAGATCTTTA AGGTGCCGGAGCCGTTTATTCCGAAATCTGGCGCGCGCGTAATGTCG CTGCTGGAGCCGACCAAGAAGATGTCCAAGTCTGACGATAATCGCAA TAACGTTATCGGCCTGCTGGAAGATCCGAAATCGGTAGTGAAGAAAA TCAAACGTGCGGTCACTGACTCCGACGAGCCGCCGGTAGTTCGCTAC GATGTGCAGAACAAAGCGGGCGTTTCCAACCTGTTGGATATCCTTTC AGCGGTAACGGGCCAGAGCATCCCAGAACTGGAAAAACAGTTCGAAG GCAAGATGTATGGTCATCTGAAAGGTGAAGTGGCTGATGCCGTTTCC GGTATGCTGACTGAATTGCAGGAACGCTATCACCGTTTCCGCAACGA TGAAGCCTTCCTGCAACAGGTGATGAAAGATGGCGCGGAAAAAGCCA GCGCGCACGCTTCCCGTACGCTAAAAGCGGTGTACGAAGCGATTGGT TTTGTGGCGAAGCCGTAA 104 GTGGGGTTCCCGAGCGGCCAAAGGGAGCAGACTtcAAATCTGCCGTC ACAGACTTCGAAGGTTCGAATCCTTCCCCCACCACCA 105 GTGGGGTTCCCGAGCGGCCAAAGGGAGCAGACTnnnAATCTGCCGTC ACAGACTTCGAAGGTTCGAATCCTTCCCCCACCACCA wherein nnn is CTA, TCA, or TTA 106 GGAAACCTGATCATGTAGATCGAACGGACTCTAAATCCGTTCAGCCG GGTTAGATTCCCGGGGTTTCCGCCA 107 GGAAACCTGATCATGTAGATCGAACGGACTnnnAATCCGTTCAGCCG GGTTAGATTCCCGGGGTTTCCGCCA 108 AGGGGCGTAGTTCAATTGGTAGAGCACCGGTCTccaAAACCGGGTGT TGGGAGTTCGAGTCTCTCCGCCCCTG 109 AGGGGCGTAGTTCAATTGGTAGAGCACCGGTCTctaAAACCGGGTGT TGGGAGTTCGAGTCTCTCCGCCCCTG 110 AGGGGCGTAGTTCAATTGGTAGAGCACCGGTCTtcaAAACCGGGTGT TGGGAGTTCGAGTCTCTCCGCCCCTG 111 AGGGGCGTAGTTCAATTGGTAGAGCACCGGTCTttaAAACCGGGTGT TGGGAGTTCGAGTCTCTCCGCCCCTG 112 TGGGGTATCGCCAAGCGGTAAGGCACCGGATTCnnnTTCCGGCATTC CGAGGTTCGAATCCTCGTACCCCAG wherein nnn is CTA, TCA, or TTA 113 AGGGGCATAGCTCAAGCGGTAAAGCACCGGACTnnnAAACCGGCAGT CCGAAGTTCGAATCCCCCCACCCCAG wherein nnn is CTA, TCA, or TTA 114 H. sapiens ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALT IgG1 SGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKV Constant DKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPE Region VTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSV LTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLP PSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG K 115 H. sapiens TVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQ Kappa SGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLS constant- SPVTKSFNRGEC region 116 VVTVPSSSLGTQTYICNVNHKPSN 117 TFGQGTKVEIKRTVAAPSVFIF

EXAMPLES

The following Examples are merely illustrative and are not intended to limit the scope or content of the invention in any way.

Example 1—Multisite Incorporation Scheme for Incorporation of Multiple Unnatural Amino Acids (UAAs) in a Protein

This Example describes the approach utilized for incorporation of multiple UAAs into a single protein.

FIG. 7 depicts the general scheme for site-specific incorporation of two distinct UAAs into proteins expressed in mammalian cells. A UGA codon is used with an appropriate tRNA/aminoacyl-tRNA synthetase (aaRS) pair 1 to incorporate a first UAA (UAA1), while a UAG codon is used with a second tRNA/aaRS pair 2 to incorporate a second UAA (UAA2). Regardless of codon location, two distinct codons in combination with appropriate tRNA/aaRS pairs will generate a ribosomally-expressed protein including two distinct UAA incorporated at their respective sites.

Selection of the components that allow for successful, efficient incorporation of multiple UAAs in a protein is challenging given that there are over two million possible combinations of distinct aaRS, tRNA, UAA, codon, and pair combinations that could be used for such multisite incorporation. As a result, it is challenging to find specific combinations of these elements that are suitable for use for site-specific incorporation of two distinct bioconjugation handles.

Example 2—TGA-Stop Codon Mediated Suppression Comparison in Mammalian Cells

This Example describes studies for analyzing effectiveness of the TGA-stop codon for incorporation of multiple UAAs into a single protein.

The following protocol was used for all small scale EGFP incorporation analyses. HEK293T cells were seeded at a density of 600,000 cells per well for a 12-well plate the day before transfection. A total amount of 1.2 μg DNA+4 μl PEI+20 μl DMEM was used for transfection of each well. For the modular three-plasmid transfection, 0.4 μg of each plasmid was used. For two-plasmid transfections, 0.6 μg of each plasmid was used. Fluorescence images were acquired by fluorescent microscopy and subsequent EGFP expression analysis were performed 48 hours post transfection. To obtain EGFP expression data, cells were harvested and lysed as described before (see PCT/US2020/045506). EGFP fluorescence in the lysate was collected in a 96-well plate using a SpectraMAX M5 (Molecular Devices) (ex=480 nm and em=530 nm). The mean of three independent experiments was reported, and error bars represent standard deviation.

For larger scale protein expression studies incorporating two UAAs, HEK293T cells were seeded in 100 mm cell culture dishes (5 million cells per dish) two days before transfection. A reaction mixture of 12 μg DNA+50 μl PEI+200 μl DMEM was used to transfect cells that were between 80%-90% confluency. The UAAs were supplemented at 1 mM final concentration at the time of transfection. Cells were harvested 48 hours post-transfection, lysed with CelLytic M, and the C-terminally polyhistidine tagged protein was purified using cobalt-containing TALON metal affinity resin columns (Clontech) following the manufacturer's protocol. Purified proteins were subjected to, for example, SDS-PAGE gels and electrospray-ionization mass spectrometry (ESI-MS) (Agilent TOF HPLC-MS) for further characterization.

FIG. 8 describes a comparison of different codon incorporation efficiencies between tRNA/aaRS pairs using pAcBac plasmids with the aminoacyl tRNA synthetase (aaRS) under a CMV promoter and tRNA under h1/U6 promoters. tRNA/aaRS used in this study were E. coli tyrosine, pyrrolysine, E. coli leucyl, and E. coli tryptophan with 1 mM OmeY, BocK, C5Az (FIG. 2, Formula 1, X═S, n=3), and HTP, respectively. Full-length EGFP reporter expression, measured by characteristic fluorescence in cell-free extract, was determined in each case to represent nonsense suppression efficiency. Each aaRS (Tyr, Pyl, Leu, or Trp) was cotransfected with either TAG or TGA suppressors of its cognate tRNA and the appropriate EGFP-mutant (TAG or TGA) to evaluate how well each pair suppressed the two different nonsense codons.

The following tRNA/aaRS pairs were used in the studies corresponding to FIG. 8: tyrosine—SEQ ID NOs: 70, 69 (CUA), and 68, pyrrolysine—SEQ ID NOs: 101, 73 (CUA), and 72, leucyl—SEQ ID NOs: 4, 16, and 17, and tryptophan—SEQ ID NOs: 45, 50, and 51.

In these studies, tryptophan displayed the highest TGA expression levels.

Example 3—Analysis of Cross Reactivity of Synthetases

This example provides an investigation of cross reactivity of selected combinations of tRNA/aaRS pairs.

An EGFP TAG or TGA reporter was used following the same protocol as described in PCT/US2020/045506. 1 mM of the UAAs OmeY, BocK, C5Az, or HTP were used, in combination with each tRNA, and with the respective LeuRS, TyrRS, PylRS, and TrpRS synthetase (FIG. 9). The following tRNA/aaRS pairs were used in the studies corresponding to FIG. 9: tyrosine—SEQ ID NOs: 70, 69 (CUA), and 68, pyrrolysine—SEQ ID NOs: 101, 73 (CUA), and 72, leucyl—SEQ ID NOs: 4, 16, and 17, and tryptophan—SEQ ID NOs: 45, 50, and 51.

FIG. 9A highlights the cross reactivity using TAG suppressor tRNAs, while FIG. 9B highlights the cross reactivity using TGA suppressor tRNAs. For example, the LeuRS only displayed selectivity towards its cognate tRNA (FIG. 9A), making LeuRS a strong candidate for use in multisite UAA incorporation; by contrast, the EcTyrRS is able to charge OmeY onto its cognate tyrosine tRNA, but was additionally cross reactive to the Leu-tRNACUA (FIG. 9A). This cross-reactive trend was also observed with TGA suppression between the tyrosyl aaRS and leucyl tRNA, albeit at a lower rate (FIG. 9B). Cross reactivity between tyrosyl and leucyl tRNA/aaRS pairs is due to the EcTyrRS, which is capable of charging the leucyl suppressor tRNAs, and therefore makes this combination incompatible for double suppression in this setting.

Example 4—Site-Specific Incorporation of Two UAAs as Demonstrated Via EGFP** Reporter Assay

This example demonstrates that it is possible to incorporate two UAAs into a protein in a mammalian cell using an EGFP reporter system.

Plasmids were constructed specifically for dual UAA incorporation. Typically, plasmid components for each pair include a CMV promoted aaRS and a multicopy cassette of tRNAs, ranging from 4-16 copies, under a U6/H1 promoter. It is contemplated that introducing the reporter under a constitutive promoter on one of the plasmids would reduce the number of plasmids needed in transfection.

The following tRNA/aaRS pairs were used in the studies corresponding to FIG. 10: tyrosine—SEQ ID NOs: 70, 69 (CUA), and 68, pyrrolysine—SEQ ID NOs: 101, 73 (CUA), and 72, leucyl—SEQ ID NOs: 4, 16, and 17, and tryptophan—SEQ ID NOs: 45, 50, and 51.

In some examples, for EcTyrTGA+PylTAG, plasmid pAcBac3-EcTyrrGA-EGFP** (EGFP** stands for EGFP-39TAG-151TGA) and pAcBac1-UbiCMbPylRS-8×tRNACUA Pyl were constructed. For EcTyrTAG+PylTGA, plasmids pAcBac3-EcTyrTAGEGFP** and pAcBac1-CMV-MbPylRS-8×tRNAUCA Pyl were constructed. pAcBac3-EcTyrTAG-EGFP** was constructed as previously described (see, for example, Zheng et al. (2017) CHEM SCI. 8(10):7211-7217), except only 16 tRNA copies were incorporated (instead of 20 copies in the original pAcBac3). The analogous pAcBac3-EcTyrTGA-EGFP** plasmid was similarly created, except 8 tRNA copies were incorporated. To generate the pAcBac1-CMV-MbPylRS-8×tRNAUCA Pyl plasmid, a pIDT-Kan-8×tRNAUCA Pyl plasmid was first prepared containing 8 copies of tandem tRNAuCA Pyl, each driven by the U6 promoter. The 8×tRNA cassette was excised out of this plasmid by NheI/AvrII digestion and inserted into the SpeI site of the previously reported pAcBac1-MbPylRS plasmid. An analogous cloning strategy was used to create the corresponding TAG-suppressing plasmid pAcBac1-UbiC-MbPylRS8×tRNACUA Pyl. For the construction of the pAcBac3-EcLeuTAG-EGFP** plasmid, a DNA sequence encoding EcLeuRS was PCR amplified to replace the AnapRS in the previously reported plasmid pAcBac2R-8×EcLtR-AnapRS3 by NheI and EcoRI restriction enzyme sites. The EcLeuRS gene was also replaced by the other aaRS variants in the below-described work. EGFP** was then PCR amplified and inserted into the SfiI sites to make the final plasmid. Similarly designed plasmids were generated for the E. coli TrpRS/tRNA pair, with minimal differences in DNA regions.

The expression levels of the multisite UAA incorporation in an EGFP** reporter were determined in FIG. 10 using a variety of combinations of pAcBac-tRNA/aaRS and pAcBac-tRNA/aaRS/EGFP**. 1 mM of each UAA was used. For example, the first plasmid combination of the tyrosine TGA suppressor and pyrrolysine TAG suppressor plasmid resulted in relatively low yields of EGFP** containing either the UAA combination OMeY+BocK or the UAA combination AzF+CpK. It is important to compare the fluorescence levels of poor but bioconjugation relevant amino acid combinations, such as AzF+CpK (797), and Cy5-N3+CpK (204), to higher yielding combinations, such as 5HTP+CpK (1226), Cy5-N3+5HTP (1077), and 5HTP+AzF (1670), which are 2-10× improved in yield compared to all other existing bioconjugation options. For the tested UAA combinations, a single band, demonstrating a homogenous product, was observed when the protein was analyzed by SDS-PAGE (FIG. 10B).

Standard whole protein ESI-MS analysis was performed for ten EGFP** variants as described above. In FIGS. 11A-G, unlabeled proteins containing two distinct UAA were analyzed for molecular weight and purity: FIG. 11A shows results of EGFP containing the UAAs HTP and BocK, FIG. 11B shows results of EGFP containing the UAAs 5HTP and OMeY, FIG. 11C shows results of EGFP containing the UAAs 5HTP and Cys-5-N3, FIG. 11D shows results of EGFP containing the UAAs 5HTP and BocK, FIG. 11E shows results of EGFP containing the UAAs 5HTP and Cyclopropene-K, FIG. 11F shows results of EGFP containing the UAAs AzW and Cyclopropene-K, and FIG. 11G shows results of EGFP containing the UAAs 5HTP and AzK. All protein samples were homogenous, demonstrating the site-specific incorporation of each UAAs. The expected mass was observed in each instance.

An EGFP** reporter contains the EGFP protein with two stop codons (TAG and TGA). Fluorophores were subsequently conjugated to the variants in one pot reactions. For click-labeling, the EGFP** protein containing an azide, 1 μg of purified EGFP** was incubated with 20 μM DBCO-Cy5 (Sigma). For EGFP** containing alkyne UAA, 1 μg protein was incubated with 50 μM Alexa Fluor 488 picolyl azide (Fisher Scientific) for 2 hours at room temperature. Identical reactions were set up with wild-type EGFP as control experiments. For labeling EGFP containing CpK, 5 μM of protein was incubated with 200 μM tetrazine-fluorescein at room temperature in PBS for 30 min. For protein containing 5HTP, 1 μg of purified EGFP** (approximately 6-10 μM) was labeled with 5× excess diazonium. FIG. 11H depicts the liquid chromatography mass spectrometry (LCMS) result of the single labeling of EGFP** containing 5HTP and AzK with a diazonium fluorophore where only the target 5HTP is labeled, using the above described conditions and depicting a single expected mass shift from the unlabeled FIG. 11G. FIG. 11I demonstrates the LCMS result of the single labeling of EGFP** containing 5HTP and AzK with DBCO-TAMRA, where only the target Azide is labeled, using the above described conditions and depicting a single expected mass shift from the unlabeled FIG. 11G. FIG. 11J demonstrates the LCMS result of the double labeling of EGFP** containing 5HTP and AzK with a diazonium fluorophore and DBCO-TAMRA, using the above described conditions and depicting a single expected mass shift from the unlabeled FIG. 11G, where 5HTP is selectively labeled with diazonium and AzK is selectively labeled with DBCO-TAMRA.

After labeling, the resulting proteins were also subjected to SDS-PAGE analysis and imaged by either the fluorescein or TAMRA settings on a BioRad Chemidoc imager. Only bands containing labeled protein are visible under these settings; the results of this analysis are shown in FIG. 12.

Example 5—Incorporation of Multiple Distinct UAA into an Antibody and Subsequent Characterization

This Example describes incorporation of two UAAs into an antibody and subsequent characterization of the antibody protein.

The ability to incorporate two distinct UAAs into a protein was extended to an antibody. The antibody utilized in this study was a full length, human IgG1 subclass monoclonal antibody (mAb) containing homologous regions to antibodies such as trastuzumab (Herceptin). A tryptophan plasmid was constructed containing a CMV promoted TrpRS.h14 (SEQ ID NO: 45) with a 4×U6 promoted Trp-tRNAUCA (SEQ ID NO: 51). A leucyl suppressor plasmid was construction containing a CMV promoted LeuRS.v1 (SEQ ID NO: 2) with a 4×U6 promoted LeutRNACUA (SEQ ID NO: 16). Light chain (LC) and heavy chain (HC) antibody fragments were expressed constitutively under a CMV promoter. It is contemplated herein that an improved system would introduce the LC and HC antibody fragments onto the suppressor plasmids to increase the transient transfection yields, which are already significantly higher than any other reported multisite suppression protein. These plasmids were transfected in Expi293 according to the manufacturer's protocols, along with one plasmid containing the HC mutants and one plasmid containing the LC mutant with a stop codon as depicted in FIG. 15.

Proteins were purified via standard Protein G columns to yield 66 mg/L and 72 mg/L total protein for HC-T202-TGA/LC-K113-TAG (incorporating the UAAs HTP and LCA) and HC-T202-TAG/LC-K113-TGA (incorporating the UAAs LCA and HTP) antibodies using 1 mM UAA as described in the manufacturer protocol and as described above. Protein yield is shown in FIG. 13A. Protein concentration was measured using the Bradford assay or Ultraviolet-visible (UV/Vis) absorbance using a Nanodrop with IgG settings. SDS-PAGE analysis (FIG. 13B) depicts clean bands for WildType (WT) (1), HTP/LCA (2) and LCA/HTP (3) with no major truncation observed. These antibodies contain two site-specific distinct UAAs with orthogonal/compatible conjugation chemistries. FIG. 14 depicts target PNGase/reduced LCMS data. The target mass of the LC containing K113-LCA is solely observed (FIG. 14A). Two peaks are observed for the HC mass (FIG. 14A). The deglycosylated target mass of 49,853 was observed, but partial de-glycosylation resulted in a glycosylation product (51,296). Without wishing to be bound by theory, this outcome may occur if the site of UAA introduction was sterically close to the glycosylation site. Regardless, importantly, a pure whole-protein mass is observed in the absence of PNGase/reduction.

HC-T202-TGA/LC-K113-TAG (HTP/LCA) antibody was labeled with diazo-PEG4-Biotin and DBCO-AF647, depicted schematically in FIG. 16, using methodologies as described above. FIG. 16 is a simplified depiction, as each labeling reagent would label the antibody twice (resulting in 4 total conjugates) due to the dimeric nature of mAbs. HPLC-HIC traces in FIG. 17 showed the appropriate shifts upon the addition of the hydrophobic molecules AF647 and biotin. FIG. 17A shows an unlabeled LC and HC peak. FIG. 17B shows the LC peak shift with the addition of DBCO-AF647. Two additional peaks are seen, but these correspond to the DBCO-AF647 additive, as clarified by a DBCO-AF647 only control, where no mAb is included (FIG. 17E) and therefore are not indicative of decreased sample purity. FIG. 17C shows the HC peak shift with the addition of diazo-biotin. FIG. 17D shows a full shift to a more hydrophobic peak when labeled together. Overall, about 100% labeling was observed by HPLC-HIC, indicating that the overall protein purity was very high.

Overall, these studies demonstrate the ability to incorporate multiple distinct UAAs into an antibody and the capability to label these antibodies (as performed with the EGFP** reporter experiments above) with therapeutic payloads or fluorophores.

INCORPORATION BY REFERENCE

The entire disclosure of each of the patent and scientific documents referred to herein is incorporated by reference for all purposes.

EQUIVALENTS

The invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting on the invention described herein. Scope of the invention is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.

Claims

1. A method of producing a protein comprising a first unnatural amino acid (UAA) and a second, different UAA, the method comprising culturing a mammalian cell with:

(i) a nucleic acid comprising a nucleotide sequence encoding a first tRNA comprising an anticodon that hybridizes to a first codon selected from UAG, UGA, and UAA, and is capable of being charged with the first UAA;
(ii) a nucleic acid comprising a nucleotide sequence encoding a first aminoacyl-tRNA synthetase capable of charging the first tRNA with the first UAA;
(iii) a nucleic acid comprising a nucleotide sequence encoding a second tRNA comprising an anticodon that hybridizes to a second codon selected from UAG, UGA, and UAA, and is capable of being charged with the second UAA, wherein the first and second tRNA do not contain the same anticodon;
(iv) a nucleic acid comprising a nucleotide sequence encoding a second aminoacyl-tRNA synthetase capable of charging the second tRNA with the second UAA; and
(v) a nucleic acid comprising a nucleotide sequence encoding the protein comprising the first codon and the second codon;
under conditions that permit the first tRNA, when expressed in the cell and charged with the first UAA, to hybridize to the first codon and direct the incorporation of the first UAA into the protein, and the second tRNA, when expressed in the cell and charged with the second UAA, to hybridize to the second codon and direct the incorporation of the second UAA into the protein;
wherein the amount of the protein comprising the first and second UAA expressed by the cell is at least 10% of the amount of a reference protein expressed by the same cell or a similar cell, wherein the reference protein is an otherwise identical protein that does not comprise the first and second UAA.

2. The method of claim 1, wherein the first tRNA is an analog or derivative of a prokaryotic tryptophanyl-tRNA.

3. The method of claim 2, wherein the prokaryotic tryptophanyl-tRNA is an E. coli tryptophanyl-tRNA.

4. The method of claim 3, wherein the first tRNA comprises a nucleotide sequence selected from any one of SEQ ID NOs: 49-54 or 108-113.

5. The method of claim 1, wherein the first aminoacyl-tRNA synthetase is an analog or derivative of a prokaryotic tryptophanyl-tRNA synthetase.

6. The method of claim 5, wherein the prokaryotic tryptophanyl-tRNA synthetase is an E. coli tryptophanyl-tRNA synthetase.

7. The method of claim 6, wherein the first aminoacyl-tRNA synthetase comprises an amino acid sequence selected from any one of SEQ ID NOs: 44-48.

8. The method of claim 1, wherein the first codon is UGA.

9. The method of claim 1, wherein the first UAA is a tryptophan analog.

10. The method of claim 9, wherein the first UAA is selected from 5-HTP and 5-AzW.

11. The method of claim 1, wherein the second tRNA is an analog or derivative of a prokaryotic leucyl-tRNA.

12. The method of claim 11, wherein the prokaryotic leucyl-tRNA is an E. coli leucyl-tRNA.

13. The method of claim 12, wherein the second tRNA comprises a nucleotide sequence selected from any one of SEQ ID NOs: 16-43.

14. The method of claim 1, wherein the second aminoacyl-tRNA synthetase is an analog or derivative of a prokaryotic leucyl-tRNA synthetase.

15. The method of claim 14, wherein the prokaryotic leucyl-tRNA synthetase is an E. coli leucyl-tRNA synthetase.

16. The method of claim 15, wherein the second aminoacyl-tRNA synthetase comprises an amino acid sequence selected from any one of SEQ ID NOs: 1-15.

17. The method of claim 1, wherein the second codon is UAG.

18. The method of claim 1, wherein the second UAA is a leucine analog.

19. The method of claim 18, wherein the second UAA is selected from LCA and Cys-5-N3.

20. The method of claim 1, wherein the second tRNA is an analog or derivative of a prokaryotic tyrosyl-tRNA.

21. The method of claim 20, wherein the prokaryotic tyrosyl-tRNA is an E. coli tyrosyl-tRNA.

22. The method of claim 21, wherein the second tRNA comprises a nucleotide sequence selected from any one of SEQ ID NOs: 68-69 or 104-105.

23. The method of claim 20, wherein the second aminoacyl-tRNA synthetase is an analog or derivative of a prokaryotic tyrosyl-tRNA synthetase.

24. The method of claim 23, wherein the prokaryotic tyrosyl-tRNA synthetase is an E. coli tyrosyl-tRNA synthetase.

25. The method of claim 24, wherein the second aminoacyl-tRNA synthetase comprises the amino acid sequence of SEQ ID NO. 70.

26. The method of claim 20, wherein the second codon is UAG.

27. The method of claim 20, wherein the second UAA is a tyrosine analog.

28. The method of claim 27, wherein the second UAA is selected from OmeY, AzF, and OpropY.

29. The method of claim 1, wherein the second tRNA is an analog or derivative of an archael pyrrolysyl-tRNA.

30. The method of claim 29, wherein the archael pyrrolysyl-tRNA is an M. barkeri pyrrolysyl-tRNA.

31. The method of claim 30, wherein the second tRNA comprises a nucleotide sequence selected from any one of SEQ ID NOs: 72-100 or 106-107.

32. The method of claim 29, wherein the second aminoacyl-tRNA synthetase is an analog or derivative of an archael pyrrolysyl-tRNA synthetase.

33. The method of claim 32, wherein the archael pyrrolysyl-tRNA synthetase is an M. barkeri pyrrolysyl-tRNA synthetase.

34. The method of claim 33, wherein the second aminoacyl-tRNA synthetase comprises the amino acid sequence of SEQ ID NO: 101.

35. The method of claim 29, wherein the second codon is UAG.

36. The method of claim 29, wherein the second UAA is a pyrrolysine analog.

37. The method of claim 36, wherein the second UAA is selected from BocK, CpK, and AzK.

38. The method of claim 1, wherein the amount of protein comprising the first and second UAA expressed by the cell is at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% of the amount of the reference protein expressed by the same cell or a similar cell.

39. The method of claim 1, wherein the reference protein comprises a wild-type amino acid residue at the positions corresponding to the first and second UAA.

40. The method of claim 1, wherein the protein is an antibody (or a fragment thereof, e.g., an antigen-binding fragment thereof), bispecific antibody, nanobody, affibody, viral protein, chemokine, cytokine, antigen, blood coagulation factor, hormone, growth factor, enzyme, cell signaling protein, or any other polypeptide or protein.

41. The method of claim 40, wherein the protein is an antibody (or a fragment thereof, e.g., an antigen-binding fragment thereof).

42. The method of claim 1, wherein the cell is a human cell.

43. The method of claim 42, wherein the cell is a human embryonic kidney (HEK) or a Chinese hamster ovary (CHO) cell.

44. The method of claim 1, further comprising purifying the protein.

45. The method of claim 1, further comprising chemically modifying the first UAA.

46. The method of claim 1, further comprising chemically modifying the second UAA.

47. The method of claim 46, wherein the chemical modification comprises conjugation to a detectable label or molecule.

48. The method of claim 47, wherein the molecule is a drug (e.g., a small molecule drug).

49. The method of claim 47, wherein the detectable label or molecule coupled to the protein via the first UAA is different from the detectable label or molecule coupled to the protein via the second UAA.

50. A method of producing a protein comprising a tryptophan analog and a leucine analog, the method comprising culturing a mammalian cell with:

(i) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA comprising an anticodon that hybridizes to a UGA codon, and is capable of being charged with the tryptophan analog;
(ii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA synthetase capable of charging the derivative of the E. coli tryptophanyl-tRNA with the tryptophan analog;
(iii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli leucyl-tRNA comprising an anticodon that hybridizes to UAG codon, and is capable of being charged with the leucine analog;
(iv) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli leucyl-tRNA synthetase capable of charging the derivative of the E. coli leucyl-tRNA with the leucine analog; and
(v) a nucleic acid comprising a nucleotide sequence encoding the protein comprising the UGA codon and the UAG codon;
under conditions that permit the derivative of the E. coli tryptophanyl-tRNA, when expressed in the cell and charged with the tryptophan analog, to hybridize to the UGA codon and direct the incorporation of the tryptophan into the protein, and the derivative of the E. coli leucyl-tRNA, when expressed in the cell and charged with the leucine analog, to hybridize to the UAG codon and direct the incorporation of the leucine analog into the protein.

51. A method of producing a protein comprising a tryptophan analog and a tyrosine analog, the method comprising culturing a mammalian cell with:

(i) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA comprising an anticodon that hybridizes to a UGA codon, and is capable of being charged with the tryptophan analog;
(ii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA synthetase capable of charging the derivative of the E. coli tryptophanyl-tRNA with the tryptophan analog;
(iii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tyrosyl-tRNA comprising an anticodon that hybridizes to UAG codon, and is capable of being charged with the tyrosine analog;
(iv) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tyrosyl-tRNA synthetase capable of charging the derivative of the E. coli tyrosyl-tRNA with the tyrosine analog; and
(v) a nucleic acid comprising a nucleotide sequence encoding the protein comprising the UGA codon and the UAG codon;
under conditions that permit the derivative of the E. coli tryptophanyl-tRNA, when expressed in the cell and charged with the tryptophan analog, to hybridize to the UGA codon and direct the incorporation of the tryptophan into the protein, and the derivative of the E. coli tyrosyl-tRNA, when expressed in the cell and charged with the tyrosine analog, to hybridize to the UAG codon and direct the incorporation of the tyrosine analog into the protein.

52. A method of producing a protein comprising a tryptophan analog and a pyrrolysine analog, the method comprising culturing a mammalian cell with:

(i) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA comprising an anticodon that hybridizes to a UGA codon, and is capable of being charged with the tryptophan analog;
(ii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an E. coli tryptophanyl-tRNA synthetase capable of charging the derivative of the E. coli tryptophanyl-tRNA with the tryptophan analog;
(iii) a nucleic acid comprising a nucleotide sequence encoding a derivative of an M. barkeri pyrrolysyl-tRNA comprising an anticodon that hybridizes to UAG codon, and is capable of being charged with the pyrrolysine analog,
(iv) a nucleic acid comprising a nucleotide sequence encoding a derivative of an M. barkeri pyrrolysyl-tRNA synthetase capable of charging the derivative of the M. barkeri pyrrolysyl-tRNA with the pyrrolysine analog; and
(v) a nucleic acid comprising a nucleotide sequence encoding the protein comprising the UGA codon and the UAG codon;
under conditions that permit the derivative of the E. coli tryptophanyl-tRNA, when expressed in the cell and charged with the tryptophan analog, to hybridize to the UGA codon and direct the incorporation of the tryptophan into the protein, and the derivative of the E. coli pyrrolysyl-tRNA, when expressed in the cell and charged with the pyrrolysine analog, to hybridize to the UAG codon and direct the incorporation of the pyrrolysine analog into the protein.

53. A protein comprising a first unnatural amino acid (UAA) and a second UAA produced by the method of claim 1.

54. A protein expressed in a mammalian cell comprising a first unnatural amino acid (UAA) that is a tryptophan analog and a second UAA that is a leucine analog.

55. The protein of claim 54, wherein the tryptophan analog is selected from 5-HTP and 5-AzW.

56. The protein of claim 54, wherein the leucine analog is selected from LCA and Cys-5-N3.

57. A protein expressed in a mammalian cell comprising a first unnatural amino acid (UAA) that is tryptophan analog and a second UAA that is tyrosine analog.

58. The protein of claim 57, wherein the tryptophan analog is selected from 5-HTP and 5-AzW.

59. The protein of claim 57, wherein the tyrosine analog is selected from OmeY, AzF, and OpropY.

60. A protein expressed in a mammalian cell comprising a first unnatural amino acid (UAA) that is a tryptophan analog and a second UAA that is a pyrrolysine analog.

61. The protein of claim 60, wherein the tryptophan analog is selected from 5-HTP and 5-AzW.

62. The protein of claim 60, wherein the pyrrolysine analog is selected from BocK, CpK, AzK, and CpK.

63. The protein of claim 53, wherein a detectable label or molecule is covalently coupled to the protein via the first UAA.

64. The protein of claim 53, wherein a detectable label or molecule is covalently coupled to the protein via the second UAA.

65. The protein of claim 64, wherein the detectable label or molecule coupled to the protein via the first UAA is different from the detectable label or molecule coupled to the protein via the second UAA.

66. The protein of claim 53, wherein the protein is an antibody (or a fragment thereof, e.g., an antigen-binding fragment thereof), bispecific antibody, nanobody, affibody, viral protein, chemokine, cytokine, antigen, blood coagulation factor, hormone, growth factor, enzyme, cell signaling protein, or any other polypeptide or protein.

67. The protein of claim 66, wherein the protein is an antibody (or a fragment thereof, e.g., an antigen-binding fragment thereof).

Patent History
Publication number: 20220403438
Type: Application
Filed: Oct 8, 2020
Publication Date: Dec 22, 2022
Inventors: Yunan Zheng (Vernon Hills, IL), James Sebastian Italia (Charlestown, MA), Abhishek Chatterjee (Lexington, MA)
Application Number: 17/767,361
Classifications
International Classification: C12P 21/02 (20060101); C12N 15/67 (20060101); C07K 14/47 (20060101); C12N 9/00 (20060101); C12N 15/11 (20060101);